US20200054711A1 - Regenerating functional neurons for treatment of neural injury caused by disruption of blood flow - Google Patents

Regenerating functional neurons for treatment of neural injury caused by disruption of blood flow Download PDF

Info

Publication number
US20200054711A1
US20200054711A1 US16/487,345 US201816487345A US2020054711A1 US 20200054711 A1 US20200054711 A1 US 20200054711A1 US 201816487345 A US201816487345 A US 201816487345A US 2020054711 A1 US2020054711 A1 US 2020054711A1
Authority
US
United States
Prior art keywords
neurod1
nucleic acid
neurons
treatment
stroke
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/487,345
Other languages
English (en)
Inventor
Gong Chen
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Penn State Research Foundation
Original Assignee
Penn State Research Foundation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Penn State Research Foundation filed Critical Penn State Research Foundation
Priority to US16/487,345 priority Critical patent/US20200054711A1/en
Publication of US20200054711A1 publication Critical patent/US20200054711A1/en
Assigned to THE PENN STATE RESEARCH FOUNDATION reassignment THE PENN STATE RESEARCH FOUNDATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHEN, GONG
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • A61K48/0058Nucleic acids adapted for tissue specific expression, e.g. having tissue specific promoters as part of a contruct
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14132Use of virus as therapeutic agent, other than vaccine, e.g. as cytolytic agent
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/30Vector systems comprising sequences for excision in presence of a recombinase, e.g. loxP or FRT
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/40Systems of functionally co-operating vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/008Vector systems having a special element relevant for transcription cell type or tissue specific enhancer/promoter combination

Definitions

  • This invention relates to compositions and methods for treating the effects of disruption of normal blood flow in the CNS of an individual subject.
  • the central nervous system (CNS) in mammals is largely unable to regenerate itself following injury.
  • Neurons and other CNS cells are often killed or injured as a result of a disease or injury which interrupts blood flow in the region where the cells are located.
  • pathological changes in the vicinity of the blood flow interruption are responsible for numerous adverse effects, such as destruction or injury of neurons, destruction or injury of glial cells, destruction or injury of blood vessels, and destruction or injury of supporting cells, in the region of the CNS affected by the disruption of normal blood flow.
  • administering exogenous NeuroD1 includes delivering an expression vector comprising a nucleic acid encoding NeuroD1 to the area.
  • administering exogenous NeuroD1 includes delivering a recombinant viral expression vector comprising a nucleic acid encoding NeuroD1 to the area.
  • NeuroD1 is the only exogenously expressed transcription factor delivered to the area.
  • Methods of treating the effects of disruption of normal blood flow in the CNS in an individual subject in need thereof are provided according to aspects of the present invention which include administering a therapeutically effective dose of exogenous NeuroD1 to an area where normal blood flow has been disrupted, wherein expressing exogenous NeuroD1 includes delivering a recombinant adeno-associated virus expression vector comprising a nucleic acid encoding NeuroD1 to the area.
  • expressing exogenous NeuroD1 includes delivering a recombinant adeno-associated virus expression vector comprising a nucleic acid encoding NeuroD1 to the area.
  • NeuroD1 is the only exogenously expressed transcription factor delivered to the area.
  • the nucleic acid sequence encoding NeuroD1 is operably linked to a glial-cell specific promoter.
  • the glial-cell specific promoter is a GFAP promoter.
  • the GFAP promoter is a human GFAP promoter.
  • Methods of treating the effects of disruption of normal blood flow in the CNS in an individual subject in need thereof are provided according to aspects of the present invention which include administering a therapeutically effective dose of exogenous NeuroD1 to an area where normal blood flow has been disrupted, wherein expressing exogenous NeuroD1 includes delivering an effective “flip-excision” recombinant expression vector combination of 1) a recombinant adeno-associated virus expression vector comprising a nucleic acid encoding NeuroD1 and 2) a recombinant adeno-associated virus expression vector comprising a nucleic acid encoding a site-specific recombinase to the area.
  • NeuroD1 is the only exogenously expressed transcription factor delivered to the area.
  • Methods of treating the effects of disruption of normal blood flow in the CNS in an individual subject in need thereof are provided according to aspects of the present invention which include administering a therapeutically effective dose of exogenous NeuroD1 to an area where normal blood flow has been disrupted, wherein expressing exogenous NeuroD1 includes delivering an effective “flip-excision” recombinant expression vector combination of 1) a recombinant adeno-associated virus expression vector comprising a nucleic acid encoding NeuroD1, wherein the nucleic acid sequence encoding NeuroD1 is operably linked to a ubiquitous promoter.
  • a recombinant adeno-associated virus expression vector comprising a nucleic acid encoding a site-specific recombinase to the area, wherein the nucleic acid sequence encoding a site-specific recombinase is operably linked to a glial-cell specific promoter.
  • the glial-cell specific promoter is a GFAP promoter.
  • the GFAP promoter is a human GFAP promoter.
  • NeuroD1 is the only exogenously expressed transcription factor delivered to the area.
  • Methods of treating the effects of disruption of normal blood flow in the CNS in an individual subject in need thereof are provided according to aspects of the present invention which include administering 1-500 ⁇ l of a pharmaceutically acceptable carrier containing adeno-associated virus particles comprising a nucleic acid encoding NeuroD1 at a concentration of 10 10 -10 14 adeno-associated virus particles/ml carrier to the subject, in the area where normal blood flow has been disrupted, at a controlled flow rate of 0.1-5 ⁇ l/min.
  • the exogenous NeuroD1 is administered at a time following disruption of normal blood flow in the CNS when reactive astrocytes are present.
  • the exogenous NeuroD1 is administered at a time following disruption of normal blood flow in the CNS when a glial scar is present.
  • the disruption of normal blood flow in the CNS is due to a disorder selected from the group consisting of: ischemia, thrombosis, embolism, hemorrhage, concussion, brain penetration, blast, inflammation, infection, tumor, chronic disease mediated restriction of blood vessels and a combination of any two or more thereof.
  • the disruption of normal blood flow in the CNS is due to a disorder selected from the group consisting of: ischemic stroke; hemorrhagic stroke; brain aneurysm; traumatic brain injury; concussion; blast; brain penetration; inflammation; infection; tumor; traumatic spinal injury; ischemic or hemorrhagic myelopathy (spinal cord infarction); global ischemia as caused by cardiac arrest or severe hypotension (shock); hypoxic-ischemic encephalopathy as caused by hypoxia, hypoglycemia, or anemia; CNS embolism as caused by infective endocarditis or atrial myxoma; fibrocartilaginous embolic myelopathy; CNS thrombosis as caused by pediatric leukemia; cerebral venous sinus thrombosis as caused by nephrotic syndrome (kidney disease), chronic inflammatory disease, pregnancy, use of estrogen-based contraceptives, meningitis, dehydration; or a combination of any disorder selected from the group consisting
  • Methods of treating the effects of disruption of normal blood flow in the CNS in an individual subject in need thereof are provided according to aspects of the present invention which include administering a therapeutically effective dose of exogenous NeuroD1 to an area where normal blood flow has been disrupted, wherein administering the therapeutically effective dose of NeuroD1 includes administering a recombinant expression vector, the expression vector including a nucleic acid sequence encoding NeuroD1 protein, wherein the nucleic acid sequence encoding NeuroD1 protein includes a nucleic acid sequence selected from the group consisting of: a nucleic acid sequence encoding SEQ ID NO:2 or a functional fragment thereof; a nucleic acid sequence encoding SEQ ID NO:4 or a functional fragment thereof; SEQ ID NO:1 or a functional fragment thereof; SEQ ID NO:3 or a functional fragment thereof; and a nucleic acid sequence encoding a protein which has 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%
  • administering the therapeutically effective dose of NeuroD1 includes stereotactic injection in or near a glial scar.
  • methods of treating the effects of disruption of normal blood flow in the CNS in an individual subject in need thereof according to the present invention further include assessing the effectiveness of the treatment in the subject.
  • methods of treating the effects of disruption of normal blood flow in the CNS in an individual subject in need thereof according to the present invention further include assessing the effectiveness of the treatment in the subject, wherein assessing the effectiveness of the treatment includes an assay selected from: electrophysiology assay, blood flow assay, tissue structure assay, function assay and a combination of any two or more thereof.
  • methods of treating the effects of disruption of normal blood flow in the CNS in an individual subject in need thereof further include assessing the effectiveness of the treatment in the subject, wherein assessing the effectiveness of the treatment includes an electroencephalogram of the subject.
  • methods of treating the effects of disruption of normal blood flow in the CNS in an individual subject in need thereof further include assessing the effectiveness of the treatment in the subject, wherein assessing the effectiveness of the treatment includes an assay of blood flow selected from the group consisting of: Near Infrared Spectroscopy and fMRI.
  • methods of treating the effects of disruption of normal blood flow in the CNS in an individual subject in need thereof further include assessing the effectiveness of the treatment in the subject, wherein assessing the effectiveness of the treatment includes an assay of tissue structure selected from the group consisting of: MRI, CAT scan, PET scan and ultrasound.
  • methods of treating the effects of disruption of normal blood flow in the CNS in an individual subject in need thereof further include assessing the effectiveness of the treatment in the subject, wherein assessing the effectiveness of the treatment includes a behavioral assay.
  • assessing the effectiveness of the treatment in the subject includes an assay performed prior to administration of the therapeutically effective dose of exogenous NeuroD1.
  • methods of treating the effects of disruption of normal blood flow in the CNS in an individual subject in need thereof include: 1) performing a first assay selected from: an electrophysiology assay, blood flow assay, tissue structure assay, function assay, or a combination of any two or more thereof, on the subject, wherein the first assay is performed prior to administration of a therapeutically effective dose of exogenous NeuroD1; 2) administering a therapeutically effective dose of exogenous NeuroD1 to an area where normal blood flow has been disrupted; and 3) performing a second assay selected from: an electrophysiology assay, blood flow assay, tissue structure assay, function assay, or a combination of any two or more thereof, on the subject, wherein the second assay is performed after administration of a therapeutically effective dose of exogenous NeuroD1.
  • compositions are provided according to aspects of the present invention which include: 1) a recombinant adeno-associated adenovirus expression vector including a glial cell specific promoter operably linked to a nucleic acid encoding a site-specific recombinase and 2) a recombinant adeno-associated adenovirus expression vector including a ubiquitous promoter operably linked to a nucleic acid encoding NeuroD1, wherein the nucleic acid encoding NeuroD1 is inverted and flanked by two sets of site-specific recombinase recognition sites such that action of the recombinase irreversibly inverts the nucleic acid encoding NeuroD1 such that NeuroD1 is expressed in a mammalian cell.
  • NeuroD1-mediated astrocyte-to-neuron conversion produced according to methods of the present invention regenerates 100-600 NeuN+new neurons/mm 2 in the stroke areas, which is 20-200 times more efficient than the internal neuroregeneration capability.
  • NeuroD1-treatment according to aspects of the present invention also decreases reactive astrocytes, reduces neuroinflammation, enhances neuronal survival, repairs blood vessels, and restores blood-brain-barrier (BBB) after stroke. Furthermore, NeuroD1-converted neurons according to aspects of the present invention not only form local neural circuits but also integrate into global brain network, achieving functional rescue of motor deficits induced by ischemic stroke. Thus, NeuroD1-mediated in vivo cell conversion according to methods of treatment of the present invention provides unprecedented neuroregenerative efficiency for the treatment of neurological disorders.
  • Methods of treatment according to aspects of the present invention demonstrate that NeuroD1-mediated in vivo glia-to-neuron conversion can functionally rescue motor deficits induced by ischemic stroke.
  • many injured neurons are preserved after reduction of reactive astrocytes and decrease of neuroinflammation.
  • NeuroD1-treatment of the present invention also repairs damaged blood vessels and restores BBB integrity in the stroke areas, leading to a landscape change from neuroinhibitory toward a neuropermissive environment.
  • Methods of treatment according to aspects of the present invention regenerate and preserve over 50-80% of the total neurons after injury resulting from disruption of normal blood flow in the CNS.
  • Brain function relies upon a delicate balance between neurons and their surrounding glial cells. After severe ischemic injury, neurons die either immediately or gradually due to the secondary injury, but their neighboring glial cells can be activated and start to proliferate, resulting in glial scar tissue that eventually inhibits neuroregeneration.
  • the NeuroD1-mediated in vivo cell conversion technology described herein restores neuronal functions in the injured areas resulting from disruption of normal blood flow in the CNS by directly converting reactive glial cells into functional neurons.
  • the in vivo glia-to-neuron conversion technology of the present invention described herein has a number of advantages compared to “classic” administration of exogenous stem cells for engraftment and attempt to treat stroke.
  • One advantage is the use endogenous glial cells instead of external cells for neuroregeneration, avoiding immunorejection associated with cell transplantation. Using endogenous glial cells that are nearby the lost neurons for regeneration is perhaps the most economical way to restore neuron functions in a local circuit. Another advantage is that the conversion of dividing reactive glial cells into non-dividing neurons not only reduces the number of reactive glial cells but also decreases the potential tumor risk associated with dividing glial cells. In contrast, “classic” stem cell therapy characterized by administration of exogenous stem cells is intrinsically linked to tumor risk. Thus, no exogenous stem cells are administered in methods of treating the effects of disruption of normal blood flow in the CNS in an individual subject in need thereof according to aspects of the present invention.
  • the NeuroD 1-mediated glia-to-neuron conversion therapy described herein can regenerate a large number of new neurons in areas characterized by disruption of normal blood flow in the CNS, such as stroke areas, averaging 400 NeuN+cells/mm 2 , which is about 100 times of the internal neurogenesis capability in the adult mouse cortex or striatum after ischemic stroke.
  • the in vivo cell conversion approach described herein makes use of reactive glial cells that are closely associated with neural injury throughout the nervous system for in situ regeneration and repair.
  • the in vivo cell conversion approach described herein also differs significantly from many other approaches currently in development for stroke treatment, which have largely focused on short-term treatment immediately after ischemic injury, such as removing blood clots, promoting blood flow, anti-inflammation, anti-oxidant, or reducing excitotoxicity. While these approaches are necessary for short-term treatment, their long-term effectiveness will be limited if a large number of neurons are lost or functionally impaired after stroke.
  • the in vivo cell conversion approach described herein offers a long-term solution by regenerating a large number of functional neurons by converting local glial cells and rebuilding local neural circuits that have been disrupted by ischemic injury.
  • the in vivo cell conversion approach described herein has a broad time window of days or weeks or months, rather than hours, after ischemic stroke for neuro-regeneration and tissue repair.
  • a short-term treatment is administered immediately after ischemic injury according to aspects of the present invention, such as removing blood clots, promoting blood flow, anti-inflammation, anti-oxidant, or reducing excitotoxicity, in addition to administering a therapeutically effective dose of exogenous NeuroD1 to an area where normal blood flow has been disrupted.
  • Reactive glial cells are a positive defense response against neural injury and killing reactive glial cells can worsen the infarct areas.
  • reactive glial cells release cytokines and inflammatory factors, including CSPG, LCN2, TNF ⁇ , IL-1 ⁇ , etc., which can inhibit axonal regeneration and neural regrowth.
  • the NeuroD 1-mediated in vivo cell conversion technology described herein turns reactive astrocytes into functional neurons, reducing the number of reactive astrocytes and the cytokines and inflammatory factors secreted by reactive glial cells.
  • astrocytes in the injury areas are replenished by newly generated astrocytes. While killing reactive astrocytes after injury results in detrimental effects, converting reactive astrocytes into neurons according to methods of treatment of the present invention yields neural repair.
  • FIGS. 1A, 1B, 1C, 1D, 1E, 1F, 1G, 1H, and 1I show establishment of a focal stroke model and NeuroD1-mediated glia-to-neuron conversion.
  • FIG. 1A shows that injection of endothelin-1 (1-31) into mouse motor cortex led to a gradual tissue loss starting from one week after the ischemic injury. Injection of PBS served as sham controls. Dashed lines indicate cortical areas. Scale bar: (A) 3 mm;
  • FIGS. 1C and FIG. 1D show results of immunostaining of NeuN (neuronal marker) and GFAP (astrocyte marker) at 5 days ( FIGS. 1C ) and 10 days post stroke (dps) ( FIGS. 1D ), respectively.
  • NeuN neurovascular marker
  • GFAP astrocyte marker
  • FIGS. 1D 10 days post stroke
  • the NeuN signal was significantly impaired in the motor cortex, and the GFAP signal was weak at 5 dps but increased significantly at 10 dps.
  • Dashed lines indicate corpus callosum.
  • Scale bar Left panel of FIGS. 1C and FIG. 1D , 200 ⁇ m, Right inset panel of FIGS. 1C and FIG. 1D 40 ⁇ m.
  • FIG. 1E shows results of injection of retroviruses carrying CAG::GFP as control or CAG::NeuroD1-IRES-GFP at 10 dps and immunostaining at 17 days post viral injection (dpi). Expressing GFP alone only labeled glial cells, whereas NeuroD1-GFP expressing cells showed immunopositive signal for NeuN. Scale bar: 20 ⁇ m.
  • FIG. 1F shows results of injection of adeno-associated virus (AAV9) carrying hGFAP::NeuroD1-P2A-GFP or hGFAP::GFP as control at 10 dps and immunostained at 17 dpi.
  • AAV9 adeno-associated virus
  • GFP control group showed mainly glial cells without NeuN signal, whereas NeuroD1 group showed many NeuN-immunopositive neurons.
  • Scale bar 40 ⁇ m.
  • FIG. 1G shows that at 4 dpi, injection of AAV9 hGFAP::Cre and CAG::FLEX-NeuroD1-P2A-mCherry resulted in significant NeuroD1 expression in GFAP-labeled astrocytes (top row). Interestingly, some NeuroD1-mCherry labeled cells showed both NeuN and GFAP signal (bottom row), suggesting a transition stage from astrocytes to neurons. Scale bar: (G) 40 ⁇ m (inset 20 ⁇ m).
  • FIG. 1H is a schematic illustration of experimental design for stroke induction, AAV injection, and immunostaining procedures. Viral injection in this scheme indicates the Cre-FLEX system carrying NeuroD1 or GFP (mCherry) control in the rest experiments in mice.
  • FIG. 1I shows that at 17 dpi, the GFP control group showed many GFAP+reactive glial cells (top row, GFAP), whereas the majority of NeuroD1-GFP labeled cells became NeuN-positive neurons (bottom row).
  • Scale bar (I) 40 ⁇ m.
  • FIGS. 2A and 2B show establishment of a focal stroke model and AAV Cre-FLEX system.
  • FIG. 2A shows representative images illustrating severe ischemic injury induced by ET-1 (1-31) in FVB mice (left), but mild injury induced by ET-1 (1-21) in FVB mice or ET-1 (1-31) in B6 mice.
  • NeuN immunostaining at 27 dps revealed that ET-1 (1-31) induced more NeuN loss and more severe cortical atrophy in FVB animals compared to the other two conditions.
  • Scale bar (A) 400 ⁇ m.
  • FIG. 2B Schematic illustration of the working mechanism of a Cre-FLEX system according to aspects of the present invention.
  • FIGS. 3A, 3B, 3C, 3D, 3E, and 3F show that NeuroD1 efficiently converts reactive astrocytes into cortical neurons.
  • FIG. 3A shows identification of astrocytes with GFAP immunostaining at 4, 7 or 17 days post viral injection (dpi) in both control and NeuroD1 groups.
  • Scale bar (A) 40 ⁇ m.
  • FIG. 3B shows identification of neurons with NeuN immunostaining at 4, 7 or 17 dpi. Arrowheads indicate some of the NeuroD1-converted neurons. Scale bar: (B) 40 ⁇ m.
  • FIG. 3E shows representative images illustrating NeuroD1-converted neurons (GFP+) expressing cortical marker (Tbr1) at 60 dpi. Note that Tbr1+cells include both converted and non-converted cells. Scale bar: (E) 40 ⁇ m.
  • FIGS. 4A, 4B, and 4C show a Tri-AAV system to trace NeuroD1-converted neurons.
  • FIG. 4A is a schematic diagram illustrating the mechanism of Tri-AAV tracing system: when only AAV-hGFAP::GFP is injected, only astrocytes will be labeled green.
  • AAV-hGFAP::GFP is injected together with the Cre-FLEX-mCherry control system, astrocytes will express both GFP and mCherry and show yellow color.
  • AAV-hGFAP::GFP is injected together with the Cre-FLEX-NeuroD1-mCherry conversion system, astrocytes will be converted into neurons and express both GFP and mCherry, appearing yellow but NeuN+.
  • FIG. 4B is a graph showing quantification of conversion efficiency by Tri-AAV tracing system at both 17 dpi and 60 dpi.
  • hGFAP::GFP group percentage of GFP+/NeuN+cells among all GFP+cells was quantified.
  • FIG. 4C shows representative images showing different levels of colocalization of GFP, mCherry and NeuN signal in three groups at 17 and 60 dpi. Scale bar: (C) 40 ⁇ m.
  • FIGS. 5A, 5B, 5C, 5D, 5E, 5F, 5G, 5H, and 5I show that NeuroD1 treatment reduces neuroinflammation in the stroke areas.
  • FIG. 5A shows representative low magnification images illustrating GFAP-labeled reactive astrocytes in the stroke cortex in control and NeuroD1 group at 17 dpi. Dashed lines indicate cortex areas. Stroke injury core is labeled by asterisk (0).Scale bar: (A) 400 ⁇ m.
  • FIG. 5B shows high magnification Images illustrating GFAP-labeled reactive astrocytes (red) in the peri-infarct areas following stroke in the GFP control group (top row) and NeuroD1 group (bottom row). Note that the astrocytes in the NeuroD1 group appeared to be less reactive in morphology compared to the control group.
  • the injury core is labeled by asterisk.
  • Scale bar (B) 40 ⁇ m.
  • FIG. 5C shows images from transgenic GFAP-GFP mice further illustrating that a significant number of astrocytes persisted in the NeuroD 1-converted areas (bottom row), suggesting that astrocytes are not depleted after cell conversion.
  • Scale bar (C) 40 ⁇ m.
  • FIG. 5D shows low magnification images showing microglia marker Iba1 in control group and NeuroD1 group at 17 dpi. Note a significant reduction of Tha1 signal in NeuroD1 group. Scale bar: (D) 400 ⁇ m.
  • FIG. 5E shows high magnification images showing reactive microglia (Iba1) with amoeboid shape in the GFP control group (top), but more ramified morphology in the NeuroD1 group (bottom).
  • Scale bar (E) 40 ⁇ m.
  • FIG. 5F shows immunofluorescent images showing a significant reduction of CSPG, an important neuroinhibitory factor released by reactive glial cells, in the NeuroD1 group compared to the control group in the peri-infarct area at 17 dpi.
  • Scale bar (F) 40 ⁇ m.
  • FIG. 5G shows immunofluorescent images showing a significant reduction of LCN2, an inflammatory factor secreted by reactive glial cells, in the NeuroD1 group compared to the control group at 17 dpi.
  • Scale bar (G) 40 ⁇ m.
  • FIG. 5I is a graph showing that RT-PCR revealed a significant increase of pro-inflammatory factors IL-1 ⁇ , IFN ⁇ , IL-6 and TNF ⁇ after stroke, but a significant decrease after NeuroD1 treatment.
  • n 4 mice for each group. Data are represented as mean ⁇ s.e.m.
  • FIGS. 6A, 6B, 6C, 6D, 6E, and 6F show reduction of reactive glial cells and inflammation after NeuroD1 treatment.
  • FIGS. 6A and FIG. 6B show representative images of astrocytes (GFAP) and microglia (Ibal) in the peri-infarct area at 7 dpi ( FIG. 6A ) and 40 dpi ( FIG. 6B ). Note that in NeuroD1 group at 40 dpi, astrocytes and microglia appeared less in number and also less reactive in morphology compared to the control group. Scale bars: 40 ⁇ m.
  • FIG. 6C Representative images of GFP (NeuroD1-GFP) co-stained with GFAP and NeuN in peri-infarct areas at 40 dpi. Note that in NeuroD1-infected areas, many astrocytes were detected and less reactive, suggesting that they were not depleted after conversion. Scale bar: 40 ⁇ m.
  • FIGS. 6D, 6E, and 6F are graphs showing quantification of the signal intensity (A.U., artificial unit) and covered area (%) for microglial marker Iba1 ( FIG. 6D ), glial scar inhibitory factor CSPG ( FIG. 6E ), and astrocyte inflammatory marker LCN2 ( FIG. 6F ).
  • One-way ANOVA followed by Tukey multiple comparison test, n 3 mice per group. Data are represented as mean ⁇ s.e.m.
  • FIGS. 7A, 7B, 7C, 7D, 7E, 7F, 7G, 7H, and 7I show that NeuroD1 regenerates new neurons and preserves injured neurons in the stroke areas.
  • FIG. 7A shows a comparison of the motor cortex (17 dpi) between the control (top row) and NeuroD1 group (bottom row).
  • Low magnification panels in the left showing the overall cortical morphology after stroke, while right panels showing enlarged images in the peri-infarct areas.
  • NeuroD1 group showed not only converted neurons (GFP+) but also non-converted neurons (NeuN+ but GFP ⁇ , arrowheads).
  • Scale bar 500 ⁇ m for the left low magnification cortex images; 40 ⁇ m for enlarged inset images;
  • FIG. 7B shows immunofluorescent staining of NeuroD1 and NeuN in NeuroD1 group at 17 dpi, illustrating both NeuroD 1-converted and non-converted neurons intermingled together. Arrowheads point to non-converted neurons (NeuN+but NeuroD1 ⁇ ). Scale bar: (B) 40 ⁇ m.
  • FIG. 7C is a graph showing quantification of NeuN+cells in peri-infarct area of control group and NeuroD1 group, with the later included both NeuroD1+ and NeuroD1-neurons. Note that the number of non-converted neurons in the NeuroD1 group more than doubled the number in the control group, suggesting a neuroprotective effect of NeuroD1 conversion.
  • FIG. 7D and FIG. 7E show immunofluorescent images of neuronal dendrite markers SMI32 ( FIG. 7D ) and MAP2 ( FIG. 7E ) showing much improved neuronal morphology in the NeuroD1 group (bottom row) compared to the control group (top row). Scale bars: 40 ⁇ m.
  • FIG. 7F and FIG. 7G show immunostaining of axonal marker SMI312 ( FIG. 7F ), NF200 ( FIG. 7G ) and axon myelination marker MBP ( FIG. 7G ) demonstrating increased axons and axonal myelination in the NeuroD1 group (bottom rows) compared to the control group (top rows). Note the better colocalization of NF200 and MBP in NeuroD1 group than the control group. Scale bars: 20 ⁇ m.
  • FIGS. 8A, 8B, 8C, 8D, and 8E show neuroregeneration and neuroprotection after NeuroD1-treatment.
  • FIG. 8A is a graph showing quantification of NeuN positive cells in the peri-infarct areas of control group and NeuroD1 group.
  • NeuroD1 group NeuN positive cells were further divided into NeuroD 1-converted (GFP+/NeuN+double positive) and non-converted neurons (GFP negative).
  • FIG. 8B shows representative images of neuronal dendritic marker Map2 in the peri-infarct area at 40 dpi. Scale bar: 40 ⁇ m.
  • FIGS. 8C, 8D, and 8E are graphs showing quantification of the signal intensity (A.U., artificial unit) and covered area (%) for dendritic marker Map2 ( FIG. 8C ), axon marker SMI312 ( FIG. 8D ) and myelination marker MBP ( FIG. 8E ).
  • One-way ANOVA followed by Tukey multiple comparison test, n 3 mice per group. Data are represented as mean ⁇ s.e.m.
  • FIGS. 9A, 9B, 9C, 9D, 9E, 9F, and 9G show reconstitution of neural circuits by NeuroD1-mediated cell conversion in the stroke areas.
  • FIG. 9A shows low magnification images illustrating cortical tissue loss in the GFP control group (top row), and the rescue of tissue loss by NeuroD1 treatment (bottom row).
  • GFP labels AAV-infected cells
  • DAPI labels cell nuclei. Dashed lines delineate the cortical areas. Scale bar: 400 ⁇ m.
  • FIG. 9B is a graph showing results of quantitative analysis of the cortical areas (from midline to 3 mm lateral) in the control versus NeuroD1 group at 7, 17, 40 and 60 dpi. Note a steady tissue loss in the control group, but a clear cortical preservation in the NeuroD1 group.
  • n 3 mice per group. Data are represented as mean ⁇ s.e.m.
  • FIG. 9C shows representative images of serial sagittal sections from medial (lower left) to lateral (upper) position, illustrating axonal projection from NeuroD1-converted neurons in the cortex (box 1) to striatum (box 2), thalamus (box 3), and hypothalamus (box 4).
  • Scale bars 1000 ⁇ m for left sagittal images; 40 ⁇ m for inset images.
  • FIG. 9D shows fluorescent (GFP) and bright field images showing NeuroD1-infected neurons in brain slices (top). Bottom, representative traces showing repetitive action potentials evoked in NeuroD1-converted neurons (60 dpi).
  • FIG. 9E shows results of Biocytin infusion in the recording pipette during whole-cell recording followed with post-fix immunostaining illustrating that the recorded neurons were both GFP+ and NeuroD1+, with complex apical dendrites at 60 dpi.
  • Scale bars 200 ⁇ m for the top low magnification image; 5 ⁇ m for inset images in bottom row.
  • FIG. 9F shows representative traces of excitatory (sEPSCs) and inhibitory synaptic events (sIPSCs) recorded in NeuroD1-GFP labeled neurons (60 dpi).
  • FIG. 9G is a graph showing quantification of the frequency of both sEPSCs and sIPSCs in cortical slices without stroke (white bar), or with stroke (black bar, GFP control; striped bar, NeuroD1 group).
  • FIGS. 10A, 10B, and 10C show brain tissue repair and long range axonal projection after NeuroD1 treatment.
  • FIG. 10A shows serial brain sections from anterior (A) to posterior (P) in the control group vs NeuroD1 group. Note severe tissue damage in the control group. ctx, cortex; c.c., corpus callosum. Scale bar: 400 ⁇ m.
  • FIG. 10B shows H&E (hematoxylin and eosin) staining at 60 dpi also shows severe tissue damage in the control group. Note an enlarged lateral ventricle in the control group, which typically associates with severe brain damage. Scale bar: 400 ⁇ m.
  • FIG. 10C shows coronal section of mouse cortex at 9 months post NeuroD1 virus injection, showing long-range axon projecting to the contralateral side through corpus callosum.
  • Bottom row illustrates enlarged view of axon projection inside the corpus callosum (ipsilateral shown in box 1 and 3, contralateral in box 4) and subcortical projection through the striatum (box 2).
  • Scale bars top row: 1000 ⁇ m, bottom row: 40 ⁇ m.
  • FIGS. 11A, 11B, 11C, 11D, 11E, 11F, 11G, and 11H show repair of blood vessels and blood-brain-barrier by NeuroD1-treatment.
  • FIG. 11A shows disruption of blood vessels and blood-brain-barrier (BBB) induced by ischemic injury.
  • Top row illustrates that in non-stroke brain, astrocytic endfeet (marked by a water channel aquaporin 4, AQP4, red) wrapped around blood vessels (labeled by monocyte and endothelial cell marker Ly6C, cyan) to form intact BBB.
  • Arrows indicate colocalization of AQP4 with GFAP around blood vessel.
  • Bottom row 1 week post stroke (wps), AQP4 signal dissociated from blood vessels, suggesting a disruption of BBB.
  • Scale bar 40 ⁇ m.
  • FIG. 11B shows comparison of blood vessels and BBB between the control and NeuroD1 groups after stroke. Top row, control group showed disrupted blood vessels and diffused AQP4 signal throughout the stroke areas. Bottom row, NeuroD1 group showed much improved blood vessels associated with AQP4 signal, suggesting repaired BBB after cell conversion (17 dpi). Scale bar: 40 ⁇ m.
  • FIG. 11C shows immunofluorescent images of another blood vessel marker CD31 costaining with AQP4, confirming the rescue of blood vessels and BBB in the NeuroD1-treated group (17 dpi).
  • Scale bar 40 ⁇ m.
  • FIG. 11D shows co-immunostaining of AQP4 and GFAP showing mislocalized AQP4 signal in reactive astrocytes in control group, compared to concentrated AQP4 signal in the endfeet of astrocytes in NeuroD1 group (17 dpi). Scale bar: 40 ⁇ m.
  • FIG. 11E shows high power images illustrating the dissociation of astrocytic endfeet (GFAP) from the blood vessels (CD31) in the control group (top), compared to a nice re-association of astrocytic endfeet with the blood vessels in NeuroD1 group (bottom).
  • Scale bar 5 ⁇ m.
  • FIGS. 11F and 11G are graphs showing quantification of AQP4 signal intensity ( FIG. 11F ) and covered area ( FIG. 11G ) in non-stroke, stroke control group, and stroke NeuroD1 group (17 dpi). NeuroD1 group is much closer to the non-stroke group. ** P ⁇ 0.01.
  • One-way ANOVA followed by Tukey multiple comparison test, n 3 mice per group. Data are represented as mean ⁇ s.e.m.
  • FIG. 11H is a graph showing quantification of blood vessels (Ly6C signal) in peri-infarct areas of control group and NeuroD1 group. NeuroD1 group showed significantly more blood vessels than the control group. **** P ⁇ 0.0001.
  • One-way ANOVA followed by Tukey multiple comparison test, n 3 per mice group. Data are represented as mean ⁇ s.e.m.
  • FIGS. 12A, 12B, 12C, 12D, 12E, 12F, 12G, 12H, and 12I show broad impact of NeuroD 1-mediated cell conversion on injured tissue, including synapse recovery and re-balance of excitation-inhibition.
  • FIG. 12A shows images of biocytin, infused during patch-clamp recording and followed with post-fix staining, showing high density of spines along the dendrites in NeuroD1-infected neurons. Scale bar: 50 ⁇ m.
  • FIG. 12B shows representative images of Golgi impregnation staining at 17 dpi showed an increase of neuronal processes and dendritic spines in the peri-infarct area of NeuroD1 group compared to controls. Scale bar: 20 ⁇ m.
  • FIG. 12E shows representative images PV-labeled interneurons in NeuroD1-converted areas and FIG. 12F is a graph showing quantification of PV-labeled interneurons in NeuroD 1-converted areas.
  • PV neurons were intermingled together with NeuroD1-converted neurons (NeuroD1-GFP) at 60 dpi. Many more PV interneurons were preserved in NeuroD1 group than in the control group. ** P ⁇ 0.01.
  • One-way ANOVA followed by Tukey multiple comparison test, n 3 animals per group. Data are represented as mean ⁇ s.e.m. Scale bar: 100 ⁇ m.
  • FIG. 12G is an illustrative image showing that in the NeuroD1 group, the AQP4 signal exhibited clear association with blood vessels in the areas with high density of NeuroD1-expressing cells but less in the neighboring areas with low density of NeuroD1-expressing cells (distinguished by dashed line). Scale bar: 40 ⁇ m.
  • FIG. 12H shows representative images of biotin perfusion analysis demonstrating that in the injury areas of control group, biotin leaked outside of the blood vessels (delineated by dashed lines, labeled by Ly6C), while biotin remained largely inside the blood vessels in the injury areas of NeuroD1 group.
  • Scale bar 5 ⁇ m.
  • FIG. 12I shows representative images of astrocytic endfeet marker AQP4 in the peri-infarct areas at 60 dpi. Note a significant re-association of AQP4 with blood vessels in NeuroD1 group. Scale bar: 40 ⁇ m.
  • FIGS. 13A, 13B, 13C, and 13D show behavioral improvement after NeuroD1-treatment.
  • FIG. 13A is a schematic illustration of experimental design for mouse forelimb motor functional tests. At 9 days post stroke (9 dps) and one day prior to viral injection, behavioral tests were performed to assess the motor functions of mice injected with ET-1 (1-31) or PBS as sham controls. AAV were injected at 10 dps, and behavioral tests were performed at the indicated time points (20, 30, 50, and 70 dps) to assess functional recovery.
  • FIG. 13B is a graph showing results of a pellet retrieval test.
  • NeuroD1 group showed accelerated recovery after stroke compared to the control group.
  • Ischemic injury at the motor cortex severely impaired the food pellet retrieval capability, from 5-6 pellets/5 min pre-stroke down to 1 pellet/5 min at 9 dps.
  • pellet retrieval ability showed steady recovery compared to the control group.
  • Note that for food pellet retrieval test only the motor cortex contralateral to the dominant side of forelimb was stroke injured and tested.
  • FIG. 13C is a graph showing results of a grid walking test.
  • NeuroD1 group showed lower foot fault rate compared to the control group. Stroke injury significantly increased the foot fault rate. NeuroD1 treatment decreased the foot fault rate.
  • FIG. 13D is a graph showing results of a cylinder test.
  • NeuroD1-treated mice showed considerable recovery of rising and touching the sidewall function of forelimbs compared to the control mice. Stroke insult impaired the mouse forelimb function in terms of rising and touching the wall of a cylinder.
  • FIGS. 14A, 14B, 14C, and 14D show assessment of tissue damage after behavioral tests.
  • FIG. 14A is a picture of the pellet retrieval device modified from staircase.
  • the mouse in the rest chamber was trained to reach the sucrose pellet in the V-shape well after 18 hr food deprivation. It is clear from this design that the mice had to spend a great effort with motor coordination and strength to successfully retrieve the food pellet.
  • FIG. 14B shows representative pictures of mouse brains after finishing behavioral tests (2 months post viral injection).
  • the control group showed severe cortical tissue loss on stroke side after ET-1 (1-31)-induced focal stroke, whereas NeuroD1-treatment showed a significant rescue of the tissue loss.
  • FIG. 14C shows representative images of the mouse cortex in coronal sections illustrating the severe tissue loss after ET-1 (1-31) induced ischemic stroke. Note that two ET-1 (1-31) injections were made in both motor cortex and sensory cortex on one side to induce more severe injury for long-lasting behavioral tests. Therefore, the tissue loss is also more severe than the single injection performed for most of the cell conversion studies. Nevertheless, the NeuroD1 repairing effect is still significant. Scale bar: 1000 ⁇ m.
  • FIG. 14D is a graph showing quantification of cortical tissue damage after behavioral tests.
  • Two-way ANOVA followed by Sidak multiple comparison test, n 6 mice per group. Data are represented as mean ⁇ s.e.m.
  • compositions and methods for treating the effects of disruption of normal blood flow in the CNS of an individual subject are provided according to aspects of the present invention.
  • a neural transcription factor NeuroD1 in reactive astrocytes treats the effects of disruption of normal blood flow in the CNS in an individual subject in need thereof.
  • methods of treatment of pathological effects of disruption of normal blood flow in the CNS of a subject including administration of a therapeutically effective amount of NeuroD1 to the subject.
  • Administration of a therapeutically effective amount of NeuroD1 to a subject affected by disruption of normal blood flow in the CNS mediates: the generation of new glutamatergic neurons by conversion of reactive astrocytes to glutamatergic neurons; reduction of the number of reactive astrocytes; survival of injured neurons including GAB Aergic and glutamatergic neurons; the generation of new non-reactive astrocytes; the reduction of reactivity of non-converted reactive astrocytes; and reintegration of blood vessels into the injured region.
  • Administration of a therapeutically effective amount of NeuroD1 to a subject affected by disruption of normal blood flow in the CNS mediates: reduced inflammation at the injury site; reduced neuroinhibition at the injury site; re-establishment of normal microglial morphology at the injury site; re-establishment of neural circuits at the injury site, increased blood vessels at the injury site; re-establishment of blood-brain-barrier at the injury site; re-establishment of normal tissue structure at the injury site; and improvement of motor deficits due to the disruption of normal blood flow.
  • the subject in need of treatment may be human or non-human mammalian, but can be non-mammalian as well.
  • administering to ameliorate the effects of disruption of normal blood flow in the CNS in an individual subject in need thereof has greater beneficial effects when administered to reactive astrocytes than to quiescent astrocytes, typically 3 to 60 days, 5 to 45 days or 8 to 30 days following the onset of disruption of normal blood flow in the CNS of the subject, although certain benefit can be achieved earlier or later, including 2 days to 1 year or later following the onset of interruption of blood flow in the CNS of the subject.
  • Administration of NeuroD1 to a subject may be used to treat injury arising from disruption of normal blood flow to the CNS, as caused by ischemia, thrombosis, embolism, hemorrhage, concussion, tumor, infection, inflammation, traumatic brain and spinal cord injury, or chronic disease mediated restriction of blood vessels, which results in neuron cell death and activation of reactive astrocytes.
  • Administration of NeuroD1 to a subject may be used to treat injury arising from disruption of normal blood flow to the CNS including, but not limited to, ischemic or hemorrhagic stroke, brain aneurysm, tumor, infection, inflammation, concussion, traumatic brain injury, traumatic spinal injury, ischemic or hemorrhagic myelopathy (spinal cord infarction), global ischemia as caused by cardiac arrest or severe hypotension (shock), hypoxic-ischemic encephalopathy as caused by hypoxia, hypoglycemia, or anemia, CNS embolism as caused by infective endocarditis or atrial myxoma, fibrocartilaginous embolic myelopathy, CNS thrombosis as caused by pediatric leukemia, cerebral venous sinus thrombosis as caused by nephrotic syndrome (kidney disease), chronic inflammatory disease, pregnancy, use of estrogen-based contraceptives, meningitis, and dehydration.
  • Methods of treatment of pathological effects of disruption of normal blood flow in the CNS of a subject including administration of a therapeutically effective amount of NeuroD1 to the subject in the region of disruption of normal blood flow in the CNS according to aspects of the present invention.
  • Methods of treatment of pathological effects of disruption of normal blood flow in the CNS of a subject including administration of a therapeutically effective amount of NeuroD1 to the subject at or near the site of disruption of normal blood flow in the CNS according to aspects of the present invention.
  • Methods of treatment of pathological effects of disruption of normal blood flow in the CNS of a subject including administration of a therapeutically effective amount of NeuroD1 to the subject in or near a glial scar caused by disruption of normal blood flow in the CNS according to aspects of the present invention.
  • NeuroD1 treatment can be administered to the region of injury as diagnosed by MRI. Electrophysiology can assess functional changes in neural firing as caused by neural cell death or injury. Non-invasive methods to assay neural damage include EEG. Disruption of blood flow to a point of injury may be non-invasively assayed via Near Infrared Spectroscopy and fMRI.
  • Blood flow within the region may either be increased, as seen in aneurysms, or decreased, as seen in ischemia.
  • Injury to the CNS caused by disruption of blood flow additionally causes short-term and long-term changes to tissue structure that can be used to diagnose point of injury. In the short term, injury will cause localized swelling. In the long term, cell death will cause points of tissue loss.
  • Non-invasive methods to assay structural changes caused by tissue death include MRI, PET scan, CAT scan, or ultrasound. These methods may be used singularly or in any combination to pinpoint the focus of injury.
  • NeuroD1 is administered at the periphery of the injury site where a glial scar will develop if the subject is untreated or where a glial scar is already present.
  • Glial scar location may be as determined by assaying tissue structure or function.
  • non-invasive methods to assay structural changes caused by tissue death include MRI, CAT scan, or ultrasound.
  • Functional assay may include EEG recording.
  • NeuroD1 is administered as an expression vector containing a DNA sequence encoding NeuroD1.
  • a viral vector including a nucleic acid encoding NeuroD1 is delivered by injection into the central or peripheral nerve tissue of a subject, such as by intracerebral injection, spinal cord injection and/or injection into the cerebrospinal fluid, or peripheral nerve ganglia.
  • Alternative viral delivery methods include but not limited to intravenous injection, intranasal infusion, intramuscle injection, intrathecal injection, and intraperitoneal injection.
  • a viral vector including a nucleic acid encoding NeuroD1 is delivered by stereotactic injection into the brain of a subject.
  • expression vector refers to a recombinant vehicle for introducing a nucleic acid encoding NeuroD1 into a host cell in vitro or in vivo where the nucleic acid is expressed to produce NeuroD1.
  • an expression vector including SEQ ID NO: 1 or 3 or a substantially identical nucleic acid sequence is expressed to produce NeuroD1 in cells containing the expression vector.
  • recombinant is used to indicate a nucleic acid construct in which two or more nucleic acids are linked and which are not found linked in nature.
  • Expression vectors include, but are not limited to plasmids, viruses, BACs and YACs. Particular viral expression vectors illustratively include those derived from adenovirus, adeno-associated virus, retrovirus, and lentivirus.
  • Method of treating a neurological condition in a subject in need thereof are provided according to aspects of the present invention which include providing a viral vector comprising a nucleic acid encoding NeuroD1; and delivering the viral vector to the central nervous system or peripheral nervous system of the subject, whereby the viral vector infects glial cells of the central nervous system or peripheral nervous system, respectively, producing infected glial cells and whereby exogenous NeuroD1 is expressed in the infected glial cells at a therapeutically effective level, wherein the expression of NeuroD1 in the infected cells results in a greater number of neurons in the subject compared to an untreated subject having the same neurological condition, whereby the neurological condition is treated.
  • the number of reactive glial cells will also be reduced, resulting in less neuroinhibitory factors released, less neuroinflammation, more blood vessels that are also evenly distributed, thereby making local environment more permissive to neuronal growth or axon penetration, hence alleviating neurological conditions.
  • Adeno-associated vectors are particularly useful in methods according to aspects of the present invention and will infect both dividing and non-dividing cells, at an injection site.
  • Adeno-associated viruses AAV are ubiquitous, noncytopathic, replication-incompetent members of ssDNA animal virus of parvoviridae family.
  • any of various recombinant adeno-associated viruses such as serotypes 1-9, can be used.
  • a “FLEX” switch approach is used to express NeuroD1 in infected cells according to aspects of the present invention.
  • the terms “FLEX” and “flip-excision” are used interchangeably to indicate a method in which two pairs of heterotypic, antiparallel 1oxP-type recombination sites are disposed on either side of an inverted NeuroD1 coding sequence which first undergo an inversion of the coding sequence followed by excision of two sites, leading to one of each orthogonal recombination site oppositely oriented and incapable of further recombination, achieving stable inversion, see for example Schnutgen et al., Nature Biotechnology 21:562-565, 2003; and Atasoy et al, J.
  • NeuroD1 is administered to a subject in need thereof by administration of 1) an adeno-associated virus expression vector including a DNA sequence encoding a site-specific recombinase under transcriptional control of an astrocyte-specific promoter such as GFAP or S100b or Aldh1L1; and 2) an adeno-associated virus expression vector including a DNA sequence encoding NeuroD1 under transcriptional control of a ubiquitous (constitutive) promoter or a neuron-specific promoter wherein the DNA sequence encoding NeuroD1 is inverted and in the wrong orientation for expression of NeuroD1 until the site-specific recombinase inverts the inverted DNA sequence encoding NeuroD1, thereby allowing expression of NeuroD1.
  • an adeno-associated virus expression vector including a DNA sequence encoding a site-specific recombinase under transcriptional control of an astrocyte-specific promoter such as GFAP or S100b or Aldh1L1
  • Site-specific recombinases and their recognition sites include, for example, Cre recombinase along with recognition sites 1oxP and 1ox2272 sites, or FLP-FRT recombination, or their combinations.
  • a concentration of 10 10 -10 14 adeno-associated virus particles/ml, 1-500 ⁇ l of volume, should be injected at a controlled flow rate of 0.1-5 ⁇ l/min.
  • an adeno-associated virus vector including a nucleic acid encoding NeuroD1 under transcriptional control of a ubiquitous (constitutive) promoter or a neuron-specific promoter wherein the DNA sequence encoding NeuroD1 is inverted and in the wrong orientation for expression of NeuroD1 and further includes sites for recombinase activity by a site specific recombinase, until the site-specific recombinase inverts the inverted DNA sequence encoding NeuroD1, thereby allowing expression of NeuroD1, is delivered by stereotactic injection into the brain of a subject along with an adeno-associated virus encoding a site specific recombinase.
  • an adeno-associated virus vector including a nucleic acid encoding NeuroD1 under transcriptional control of a ubiquitous (constitutive) promoter or a neuron-specific promoter wherein the DNA sequence encoding NeuroD1 is inverted and in the wrong orientation for expression of NeuroD1 and further includes sites for recombinase activity by a site specific recombinase, until the site-specific recombinase inverts the inverted DNA sequence encoding NeuroD1, thereby allowing expression of NeuroD1, is delivered by stereotactic injection into the brain of a subject along with an adeno-associated virus encoding a site specific recombinase in the region of or at the site of disruption of normal blood flow in the CNS according to aspects of the present invention.
  • the site of stereotactic injection is in or near a glial scar caused by disruption of normal blood flow in the CNS.
  • the site-specific recombinase is Cre recombinase and the sites for recombinase activity are recognition sites 1oxP and 1ox2272 sites.
  • NeuroD1 treatment of a subject is monitored during or after treatment to monitor progress and/or final outcome of the treatment.
  • Post-Treatment Assay for successful neuronal cell integration and restoration of tissue microenvironment is diagnosed by restoration or near-restoration of normal electrophysiology, blood flow, tissue structure, and function.
  • Non-invasive methods to assay neural function include EEG. Blood flow may be non-invasively assayed via Near Infrared Spectroscopy and fMRI.
  • Non-invasive methods to assay tissue structure include MRI, CAT scan, PET scan, or ultrasound.
  • Behavioral assays may be used to non-invasively assay for restoration of CNS function. The behavioral assay should be matched to the loss of function caused by original CNS injury.
  • Assays to evaluate treatment may be performed at any point, such as 1 day, 2 days, 3 days, one week, 2 weeks, 3 weeks, one month, or later, after NeuroD1 treatment. Such assays may be performed prior to NeuroD1 treatment in order to establish a baseline comparison if desired.
  • RNA Interference Nuts and Bolts of RNAi Technology, DNA Press LLC, Eagleville, Pa., 2003; Herdewijn, p. (Ed.), Oligonucleotide Synthesis: Methods and Applications, Methods in Molecular Biology, Humana Press, 2004; A. Nagy, M. Gertsenstein, K. Vintersten, R. Behringer, Manipulating the Mouse Embryo: A Laboratory Manual, Cold Spring Harbor Laboratory Press,3 rd Ed.; Dec.
  • NeuroD1 protein refers to a bHLH proneural transcription factor involved in embryonic brain development and in adult neurogenesis, see Cho, J. H. et al., Mol, Neurobiol., 30:35-47, 2004; Kuwabara, T. et al., Nature Neurosci., 12:1097-1105, 2009; and Gao, Z. et al., Nature Neurosci., 12:1090-1092, 2009. NeuroD1 is expressed late in development, mainly in the nervous system and is involved in neuronal differentiation, maturation and survival.
  • NeuroD1 protein encompasses human NeuroD1 protein, identified here as SEQ ID NO: 2 and mouse NeuroD1 protein, identified here as SEQ ID NO: 4.
  • NeuroD1 protein encompasses variants of NeuroD1 protein, such as variants of SEQ ID NO: 2 and SEQ ID NO: 4, which may be included in methods of the present invention.
  • variant refers to naturally occurring genetic variations and recombinantly prepared variations, each of which contain one or more changes in its amino acid sequence compared to a reference NeuroD1 protein, such as SEQ ID NO: 2 or SEQ ID NO: 4.
  • Such changes include those in which one or more amino acid residues have been modified by amino acid substitution, addition or deletion.
  • the term “variant” encompasses orthologs of human NeuroD1, including for example mammalian and bird NeuroD1, such as, but not limited to NeuroD1 orthologs from a non-human primate, cat, dog, sheep, goat, horse, cow, pig, bird, poultry animal and rodent such as but not limited to mouse and rat.
  • mouse NeuroD1 exemplified herein as amino acid sequence SEQ ID NO: 4 is an ortholog of human NeuroD1.
  • Preferred variants have at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 2 or SEQ ID NO: 4.
  • Mutations can be introduced using standard molecular biology techniques, such as site-directed mutagenesis and PCR-mediated mutagenesis.
  • One of skill in the art will recognize that one or more amino acid mutations can be introduced without altering the functional properties of the NeuroD1 protein.
  • one or more amino acid substitutions, additions, or deletions can be made without altering the functional properties of the NeuroD1 protein of SEQ ID NO: 2 or 4.
  • Conservative amino acid substitutions can be made in a NeuroD1 protein to produce a NeuroD1 protein variant.
  • Conservative amino acid substitutions are art recognized substitutions of one amino acid for another amino acid having similar characteristics.
  • each amino acid may be described as having one or more of the following characteristics: electropositive, electronegative, aliphatic, aromatic, polar, hydrophobic and hydrophilic.
  • a conservative substitution is a substitution of one amino acid having a specified structural or functional characteristic for another amino acid having the same characteristic.
  • Acidic amino acids include aspartate, glutamate; basic amino acids include histidine, lysine, arginine; aliphatic amino acids include isoleucine, leucine and valine; aromatic amino acids include phenylalanine, glycine, tyrosine and tryptophan; polar amino acids include aspartate, glutamate, histidine, lysine, asparagine, glutamine, arginine, serine, threonine and tyrosine; and hydrophobic amino acids include alanine, cysteine, phenylalanine, glycine, isoleucine, leucine, methionine, proline, valine and tryptophan; and conservative substitutions include substitution among amino acids within each group. Amino acids may also be described in terms of relative size, alanine, cysteine, aspartate, glycine, asparagine, proline, threonine, serine, valine, all typically considered to be small.
  • NeuroD1 variants can include synthetic amino acid analogs, amino acid derivatives and/or non-standard amino acids, illustratively including, without limitation, alpha-aminobutyric acid, citrulline, canavanine, cyanoalanine, diaminobutyric acid, diaminopimelic acid, dihydroxy-phenylalanine, djenkolic acid, homoarginine, hydroxyproline, norleucine, norvaline, 3-phosphoserine, homoserine, 5-hydroxytryptophan, 1-methylhistidine, 3-methylhistidine, and ornithine.
  • synthetic amino acid analogs amino acid derivatives and/or non-standard amino acids
  • amino acid derivatives illustratively including, without limitation, alpha-aminobutyric acid, citrulline, canavanine, cyanoalanine, diaminobutyric acid, diaminopimelic acid, dihydroxy-phenylalanine, djenkolic acid, homoarginine, hydroxy
  • the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in the sequence of a first amino acid or nucleic acid sequence for optimal alignment with a second amino acid or nucleic acid sequence).
  • the amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position.
  • the determination of percent identity between two sequences can also be accomplished using a mathematical algorithm.
  • a preferred, non-limiting example of a mathematical algorithm utilized for the comparison of two sequences is the algorithm of Karlin and Altschul, 1990, PNAS 87:2264 2268, modified as in Karlin and Altschul, 1993, PNAS. 90:5873 5877. Such an algorithm is incorporated into the NBLAST and XBLAST programs of Altschul et al., 1990, J. Mol. Biol. 215:403.
  • Gapped BLAST are utilized as described in Altschul et al., 1997, Nucleic Acids Res. 25:3389 3402.
  • PSI BLAST is used to perform an iterated search which detects distant relationships between molecules.
  • the default parameters of the respective programs e.g., of XBLAST and NBLAST
  • the default parameters of the respective programs are used (see, e.g., the NCBI website).
  • a mathematical algorithm utilized for the comparison of sequences is the algorithm of Myers and Miller, 1988, CABIOS 4:11 17. Such an algorithm is incorporated in the ALIGN program (version 2.0) which is part of the GCG sequence alignment software package. When utilizing the ALIGN program for comparing amino acid sequences, a PAM120 weight residue table, a gap length penalty of 12, and a gap penalty of 4 is used.
  • the percent identity between two sequences is determined using techniques similar to those described above, with or without allowing gaps. In calculating percent identity, typically only exact matches are counted.
  • NeuroD1 protein encompasses fragments of the NeuroD1 protein, such as fragments of SEQ ID NOs. 2 and 4 and variants thereof, operable in methods and compositions of the present invention.
  • NeuroD1 proteins and nucleic acids may be isolated from natural sources, such as the brain of an organism or cells of a cell line which expresses NeuroD1.
  • NeuroD1 protein or nucleic acid may be generated recombinantly, such as by expression using an expression construct, in vitro or in vivo.
  • NeuroD1 proteins and nucleic acids may also be synthesized by well-known methods.
  • NeuroD1 included in methods and compositions of the present invention is preferably produced using recombinant nucleic acid technology.
  • Recombinant NeuroD1 production includes introducing a recombinant expression vector encompassing a DNA sequence encoding NeuroD1 into a host cell.
  • a nucleic acid sequence encoding NeuroD1 introduced into a host cell to produce NeuroD1 encodes SEQ ID NO: 2, SEQ ID NO: 4, or a variant thereof.
  • the nucleic acid sequence identified herein as SEQ ID NO: 1 encodes SEQ ID NO: 2 and is included in an expression vector and expressed to produce NeuroD1.
  • the nucleic acid sequence identified herein as SEQ ID NO: 3 encodes SEQ ID NO: 4 and is included in an expression vector and expressed to produce NeuroD1.
  • nucleic acid sequences substantially identical to SEQ ID NOs. 1 and 3 encode NeuroD1 and variants of NeuroD1, and that such alternate nucleic acids may be included in an expression vector and expressed to produce NeuroD1 and variants of NeuroD1.
  • a fragment of a nucleic acid encoding NeuroD1 protein can be used to produce a fragment of a NeuroD1 protein.
  • An expression vector contains a nucleic acid that includes segment encoding a polypeptide of interest operably linked to one or more regulatory elements that provide for transcription of the segment encoding the polypeptide of interest.
  • operably linked refers to a nucleic acid in functional relationship with a second nucleic acid.
  • operably linked encompasses functional connection of two or more nucleic acid molecules, such as a nucleic acid to be transcribed and a regulatory element.
  • regulatory element refers to a nucleotide sequence which controls some aspect of the expression of an operably linked nucleic acid.
  • Exemplary regulatory elements include an enhancer, such as, but not limited to: woodchuck hepatitis virus posttranscriptional regulatory element (WPRE); an internal ribosome entry site (IRES) or a 2A domain; an intron; an origin of replication; a polyadenylation signal (pA); a promoter; a transcription termination sequence; and an upstream regulatory domain, which contribute to the replication, transcription, post-transcriptional processing of an operably linked nucleic acid sequence.
  • WPRE woodchuck hepatitis virus posttranscriptional regulatory element
  • IVS internal ribosome entry site
  • promoter refers to a DNA sequence operably linked to a nucleic acid sequence to be transcribed such as a nucleic acid sequence encoding NeuroD1.
  • a promoter is generally positioned upstream of a nucleic acid sequence to be transcribed and provides a site for specific binding by RNA polymerase and other transcription factors.
  • a promoter is generally positioned upstream of the nucleic acid sequence transcribed to produce the desired molecule, and provides a site for specific binding by RNA polymerase and other transcription factors.
  • the 5′ non-coding region of a gene can be isolated and used in its entirety as a promoter to drive expression of an operably linked nucleic acid.
  • a portion of the 5′ non-coding region can be isolated and used to drive expression of an operably linked nucleic acid.
  • about 500-6000 bp of the 5′ non-coding region of a gene is used to drive expression of the operably linked nucleic acid.
  • a portion of the 5′ non-coding region of a gene containing a minimal amount of the 5′ non-coding region needed to drive expression of the operably linked nucleic acid is used.
  • Assays to determine the ability of a designated portion of the 5′ non-coding region of a gene to drive expression of the operably linked nucleic acid are well-known in the art.
  • promoters used to drive expression of NeuroD1 are “ubiquitous” or “constitutive” promoters, that drive expression in many, most, or all cell types of an organism into which the expression vector is transferred.
  • ubiquitous promoters that can be used in expression of NeuroD1 are cytomegalovirus promoter; simian virus 40 (SV40) early promoter; rous sarcoma virus promoter; adenovirus major late promoter; beta actin promoter; glyceraldehyde 3-phosphate dehydrogenase; glucose-regulated protein 78 promoter; glucose-regulated protein 94 promoter; heat shock protein 70 promoter; beta-kinesin promoter; ROSA promoter; ubiquitin B promoter; eukaryotic initiation factor 4A1 promoter and elongation Factor I promoter; all of which are well-known in the art and which can be isolated from primary sources using routine methodology or obtained from commercial sources. Promoters can be derived entirely from a simian virus 40 (SV40
  • Combinations of regulatory sequences may be included in an expression vector and used to drive expression of NeuroD1.
  • a non-limiting example included in an expression vector to drive expression of NeuroD1 is the CAG promoter which combines the cytomegalovirus CMV early enhancer element and chicken beta-actin promoter.
  • promoters used to drive expression of NeuroD1 according to methods described herein are those that drive expression preferentially in glial cells, particularly astrocytes and/or NG2 cells. Such promoters are termed “astrocyte-specific” and/or “NG2 cell-specific” promoters.
  • Non-limiting examples of astrocyte-specific promoters are glial fibrillary acidic protein (GFAP) promoter and aldehyde dehydrogenase 1 family, member L1 (Aldh1L1) promoter.
  • GFAP glial fibrillary acidic protein
  • Aldh1L1 aldehyde dehydrogenase 1 family, member L1
  • Human GFAP promoter is shown herein as SEQ ID NO:6
  • Mouse Aldh1L1 promoter is shown herein as SEQ ID NO:7.
  • NG2 cell-specific promoter is the promoter of the chondroitin sulfate proteoglycan 4 gene, also known as neuron-glial antigen 2 (NG2).
  • NG2 promoter is shown herein as SEQ ID NO:8.
  • promoters used to drive expression of NeuroD1 according to methods described herein are those that drive expression preferentially in reactive glial cells, particularly reactive astrocytes and/or reactive NG2 cells. Such promoters are termed “reactive astrocyte-specific” and/or “reactive NG2 cell-specific” promoters.
  • a non-limiting example of a “reactive astrocyte-specific” promoter is the promoter of the lipocalin 2 (1cn2) gene.
  • Mouse 1cn2 promoter is shown herein as SEQ ID NO:5.
  • Homologues and variants of ubiquitous and cell type-specific promoters may be used in expressing NeuroD1.
  • Promoter homologues and promoter variants can be included in an expression vector for expressing NeuroD1 according to the present invention.
  • the terms “promoter homologue” and “promoter variant” refer to a promoter which has substantially similar functional properties to confer the desired type of expression, such as cell type-specific expression of NeuroD1 or ubiquitous expression of NeuroD1, on an operably linked nucleic acid encoding NeuroD1 compared to those disclosed herein.
  • a promoter homologue or variant has substantially similar functional properties to confer cell type-specific expression on an operably linked nucleic acid encoding NeuroD1 compared to GFAP, S100b, Aldh1L1, NG2, 1cn2 and CAG promoters.
  • promoter variant refers to either an isolated naturally occurring or a recombinantly prepared variation of a reference promoter, such as, but not limited to, GFAP, S100b, Aldh1L1, NG2, 1cn2 and pCAG promoters.
  • promoters from other species are functional, e.g. the mouse Aldh1L1 promoter is functional in human cells.
  • Homologues and homologous promoters from other species can be identified using bioinformatics tools known in the art, see for example, Xuan et al., 2005, Genome Biol 6:R72; Zhao et al., 2005, Nucl Acid Res 33:D103-107; and Halees et al. 2003, Nucl. Acids. Res. 2003 31: 3554-3559.
  • homologues and variants of cell type-specific promoters of NeuroD1 or and/or ubiquitous promoters have at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or greater, nucleic acid sequence identity to the reference developmentally regulated and/or ubiquitous promoter and include a site for binding of RNA polymerase and, optionally, one or more binding sites for transcription factors.
  • a nucleic acid sequence which is substantially identical to SEQ ID NO:1 or SEQ ID NO:3 is characterized as having a complementary nucleic acid sequence capable of hybridizing to SEQ ID NO:1 or SEQ ID NO:3 under high stringency hybridization conditions.
  • nucleic acid sequences encoding additional proteins can be included in an expression vector.
  • additional proteins include non-NeuroD1 proteins such as reporters, including, but not limited to, beta-galactosidase, green fluorescent protein and antibiotic resistance reporters.
  • the recombinant expression vector encodes at least NeuroD1 of SEQ ID NO:2, a protein having at least 95% identity to SEQ ID NO:2, or a protein encoded by a nucleic acid sequence substantially identical to SEQ ID NO:1.
  • the recombinant expression vector encodes at least NeuroD1 of SEQ ID NO:4, a protein having at least 95% identity to SEQ ID NO:4, or a protein encoded by a nucleic acid sequence substantially identical to SEQ ID NO:2.
  • SEQ ID NO:9 is an example of a nucleic acid including CAG promoter operably linked to a nucleic acid encoding NeuroD1, and further including a nucleic acid sequence encoding EGFP and an enhancer, WPRE.
  • An IRES separates the nucleic acid encoding NeuroD1 and the nucleic acid encoding EGFP.
  • SEQ ID NO:9 is inserted into an expression vector for expression of NeuroD1 and the reporter gene EGFP.
  • the IRES and nucleic acid encoding EGFP are removed and the remaining CAG promoter and operably linked nucleic acid encoding NeuroD1 is inserted into an expression vector for expression of NeuroD1.
  • the WPRE or another enhancer is optionally included.
  • a reporter gene is included in a recombinant expression vector encoding NeuroD1.
  • a reporter gene may be included to produce a peptide or protein that serves as a surrogate marker for expression of NeuroD1 from the recombinant expression vector.
  • reporter gene refers to gene that is easily detectable when expressed, for example by chemiluminescence, fluorescence, colorimetric reactions, antibody binding, inducible markers and/or ligand binding assays.
  • Exemplary reporter genes include, but are not limited to, green fluorescent protein (GFP), enhanced green fluorescent protein (eGFP), yellow fluorescent protein (YFP), enhanced yellow fluorescent protein (eYFP), cyan fluorescent protein (CFP), enhanced cyan fluorescent protein (eCFP), blue fluorescent protein (BFP), enhanced blue fluorescent protein (eBFP), MmGFP (Zernicka-Goetz et al., Development, 124:1133-1137, 1997, dsRed, luciferase and beta-galactosidase (lacZ).
  • GFP green fluorescent protein
  • eGFP enhanced green fluorescent protein
  • YFP yellow fluorescent protein
  • eYFP enhanced yellow fluorescent protein
  • CFP cyan fluorescent protein
  • BFP blue fluorescent protein
  • eBFP enhanced blue fluorescent protein
  • MmGFP Zernicka-Goetz et al., Development, 124:1133-1137, 1997, dsRed, luciferase and beta-galactosidase (lacZ).
  • transfection The process of introducing genetic material into a recipient host cell, such as for transient or stable expression of a desired protein encoded by the genetic material in the host cell is referred to as “transfection.”
  • Transfection techniques are well-known in the art and include, but are not limited to, electroporation, particle accelerated transformation also known as “gene gun” technology, liposome-mediated transfection, calcium phosphate or calcium chloride co-precipitation-mediated transfection, DEAE-dextran-mediated transfection, microinjection, polyethylene glycol mediated transfection, heat shock mediated transfection and virus-mediated transfection.
  • virus-mediated transfection may be accomplished using a viral vector such as those derived from adenovirus, adeno-associated virus and lentivirus.
  • a host cell is transfected ex-vivo and then re-introduced into a host organism.
  • cells or tissues may be removed from a subject, transfected with an expression vector encoding NeuroD1 and then returned to the subject.
  • a recombinant expression vector including a nucleic acid encoding NeuroD1, or a functional fragment thereof, into a host glial cell in vitro or in vivo for expression of exogenous NeuroD1 in the host glial cell to convert the glial cell to a neuron is accomplished by any of various transfection methodologies.
  • exogenous NeuroD1 in the host glial cell to convert the glial cell to a neuron is optionally achieved by introduction of mRNA encoding NeuroD1, or a functional fragment thereof, to the host glial cell in vitro or in vivo.
  • An expression vector including a nucleic acid encoding NeuroD1 or a functional fragment thereof, mRNA encoding NeuroD1 or a functional fragment thereof, and/or NeuroD1 protein, full-length or a functional fragment thereof, is optionally associated with a carrier for introduction into a host cell in vitro or in vivo.
  • the carrier is a particulate carrier such as lipid particles including liposomes, micelles, unilamellar or mulitlamellar vesicles; polymer particles such as hydrogel particles, polyglycolic acid particles or polylactic acid particles; inorganic particles such as calcium phosphate particles such as described in for example U.S. Pat. No. 5,648,097; and inorganic/organic particulate carriers such as described for example in U.S. Pat. No. 6,630,486.
  • lipid particles including liposomes, micelles, unilamellar or mulitlamellar vesicles
  • polymer particles such as hydrogel particles, polyglycolic acid particles or polylactic acid particles
  • inorganic particles such as calcium phosphate particles such as described in for example U.S. Pat. No. 5,648,097
  • inorganic/organic particulate carriers such as described for example in U.S. Pat. No. 6,630,486.
  • a particulate carrier can be selected from among a lipid particle; a polymer particle;
  • an inorganic particle and an inorganic/organic particle.
  • a mixture of particle types can also be included as a particulate pharmaceutically acceptable carrier.
  • a particulate carrier is typically formulated such that particles have an average particle size in the range of about 1 nm-10 microns.
  • a particulate carrier is formulated such that particles have an average particle size in the range of about 1 nm-100 nm.
  • liposomes and methods relating to their preparation and use may be found in Liposomes: A Practical Approach (The Practical Approach Series, 264), V. P. Torchilin and V. Weissig (Eds.), Oxford University Press; 2nd ed., 2003. Further aspects of nanoparticles are described in S. M. Moghimi et al., FASEB J. 2005, 19, 311-30.
  • Expression of NeuroD1 using a recombinant expression vector is accomplished by introduction of the expression vector into a eukaryotic or prokaryotic host cell expression system such as an insect cell, mammalian cell, yeast cell, bacterial cell or any other single or multicellular organism recognized in the art.
  • Host cells are optionally primary cells or immortalized derivative cells.
  • Immortalized cells are those which can be maintained in-vitro for at least 5 replication passages.
  • Host cells containing the recombinant expression vector are maintained under conditions wherein NeuroD1 is produced.
  • Host cells may be cultured and maintained using known cell culture techniques such as described in Celis, Julio, ed., 1994, Cell Biology Laboratory Handbook, Academic Press, N.Y.
  • Various culturing conditions for these cells including media formulations with regard to specific nutrients, oxygen, tension, carbon dioxide and reduced serum levels, can be selected and optimized by one of skill in the art.
  • a recombinant expression vector including a nucleic acid encoding NeuroD1 is introduced into glial cells of a subject. Expression of exogenous NeuroD1 in the glial cells “converts” the glial cells into neurons.
  • a recombinant expression vector including a nucleic acid encoding NeuroD1 or a functional fragment thereof is introduced into astrocytes of a subject. Expression of exogenous NeuroD1 in the glial cells “converts” the astrocytes into neurons.
  • a recombinant expression vector including a nucleic acid encoding NeuroD1 or a functional fragment thereof is introduced into reactive astrocytes of a subject. Expression of exogenous NeuroD1 or a functional fragment thereof in the reactive astrocytes “converts” the reactive astrocytes into neurons.
  • a recombinant expression vector including a nucleic acid encoding NeuroD1 or a functional fragment thereof is introduced into NG2 cells of a subject. Expression of exogenous NeuroD1 or a functional fragment thereof in the NG2 cells “converts” the NG2 cells into neurons.
  • Detection of expression of exogenous NeuroD1 following introduction of a recombinant expression vector including a nucleic acid encoding the exogenous NeuroD1 or a functional fragment thereof is accomplished using any of various standard methodologies including, but not limited to, immunoassays to detect NeuroD1, nucleic acid assays to detect NeuroD1 nucleic acids and detection of a reporter gene co-expressed with the exogenous NeuroD1.
  • neuroD1 converted neurons and “converted neurons” are used herein to describe the effect of expression of NeuroD1 or a functional fragment thereof resulting in a change of a glial cell, astrocyte or reactive astrocyte phenotype to a neuronal phenotype.
  • Neuronal phenotype a glial cell, astrocyte or reactive astrocyte phenotype to a neuronal phenotype.
  • Neuronal phenotype a cell including exogenous NeuroD1 protein or a functional fragment thereof which has consequent neuronal phenotype.
  • neuronal phenotype refers to well-known detectable characteristics of the cells referred to herein.
  • the neuronal phenotype can be, but is not limited to, one or more of: neuronal morphology, expression of one or more neuronal markers, electrophysiological characteristics of neurons, synapse formation and release of neurotransmitter.
  • neuronal phenotype encompasses but is not limited to: characteristic morphological aspects of a neuron such as presence of dendrites, an axon and dendritic spines; characteristic neuronal protein expression and distribution, such as presence of synaptic proteins in synaptic puncta, presence of MAP2 in dendrites; and characteristic electrophysiological signs such as spontaneous and evoked synaptic events.
  • glial phenotype such as astrocyte phenotype and reactive astrocyte phenotypes encompasses but is not limited to: characteristic morphological aspects of astrocytes and reactive astrocytes such as a generally “star-shaped” morphology; and characteristic astrocyte and reactive astrocyte protein expression, such as presence of glial fibrillary acidic protein (GFAP).
  • characteristic morphological aspects of astrocytes and reactive astrocytes such as a generally “star-shaped” morphology
  • characteristic astrocyte and reactive astrocyte protein expression such as presence of glial fibrillary acidic protein (GFAP).
  • GFAP glial fibrillary acidic protein
  • nucleic acid refers to RNA or DNA molecules having more than one nucleotide in any form including single-stranded, double-stranded, oligonucleotide or polynucleotide.
  • nucleotide sequence refers to the ordering of nucleotides in an oligonucleotide or polynucleotide in a single-stranded form of nucleic acid.
  • NeuroD1 nucleic acid refers to an isolated NeuroD1 nucleic acid molecule and encompasses isolated NeuroD1 nucleic acids having a sequence that has at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the DNA sequence set forth in SEQ ID NO:1 or SEQ ID NO:3, or the complement thereof, or a fragment thereof, or an isolated DNA molecule having a sequence that hybridizes under high stringency hybridization conditions to the nucleic acid set forth as SEQ ID NO:1 or SEQ ID NO:3, a complement thereof or a fragment thereof.
  • the nucleic acid of SEQ ID NO:3 is an example of an isolated DNA molecule having a sequence that hybridizes under high stringency hybridization conditions to the nucleic acid set forth in SEQ ID NO:1.
  • a fragment of a NeuroD1 nucleic acid is any fragment of a NeuroD1 nucleic acid that is operable in aspects of the present invention including a NeuroD1 nucleic acid.
  • a nucleic acid probe or primer able to hybridize to a target NeuroD1 mRNA or cDNA can be used for detecting and/or quantifying mRNA or cDNA encoding NeuroD1 protein.
  • a nucleic acid probe can be an oligonucleotide of at least 10, 15, 30, 50 or 100 nucleotides in length and sufficient to specifically hybridize under stringent conditions to NeuroD1 mRNA or cDNA or complementary sequence thereof.
  • a nucleic acid primer can be an oligonucleotide of at least 10, 15 or 20 nucleotides in length and sufficient to specifically hybridize under stringent conditions to the mRNA or cDNA, or complementary sequence thereof.
  • nucleic acid refers to Watson-Crick base pairing between nucleotides and specifically refers to nucleotides hydrogen bonded to one another with thymine or uracil residues linked to adenine residues by two hydrogen bonds and cytosine and guanine residues linked by three hydrogen bonds.
  • a nucleic acid includes a nucleotide sequence described as having a “percent complementarity” to a specified second nucleotide sequence.
  • a nucleotide sequence may have 80%, 90%, or 100% complementarity to a specified second nucleotide sequence, indicating that 8 of 10, 9 of 10 or 10 of 10 nucleotides of a sequence are complementary to the specified second nucleotide sequence.
  • the nucleotide sequence 3′-TCGA-5′ is 100% complementary to the nucleotide sequence 5′-AGCT-3′.
  • the nucleotide sequence 3′-TCGA- is 100% complementary to a region of the nucleotide sequence 5′-TTAGCTGG-3′.
  • hybridization and “hybridizes” refer to pairing and binding of complementary nucleic acids. Hybridization occurs to varying extents between two nucleic acids depending on factors such as the degree of complementarity of the nucleic acids, the melting temperature, Tm, of the nucleic acids and the stringency of hybridization conditions, as is well known in the art.
  • stringency of hybridization conditions refers to conditions of temperature, ionic strength, and composition of a hybridization medium with respect to particular common additives such as formamide and Denhardt's solution.
  • hybridization conditions relating to a specified nucleic acid are routine and is well known in the art, for instance, as described in J. Sambrook and D. W. Russell, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press; 3rd Ed., 2001; and F. M. Ausubel, Ed., Short Protocols in Molecular Biology, Current Protocols; 5th Ed., 2002.
  • High stringency hybridization conditions are those which only allow hybridization of substantially complementary nucleic acids. Typically, nucleic acids having about 85-100% complementarity are considered highly complementary and hybridize under high stringency conditions.
  • Intermediate stringency conditions are exemplified by conditions under which nucleic acids having intermediate complementarity, about 50-84% complementarity, as well as those having a high degree of complementarity, hybridize.
  • low stringency hybridization conditions are those in which nucleic acids having a low degree of complementarity hybridize.
  • specific hybridization and “specifically hybridizes” refer to hybridization of a particular nucleic acid to a target nucleic acid without substantial hybridization to nucleic acids other than the target nucleic acid in a sample.
  • Hybridization and conditions to achieve a desired hybridization stringency are described, for example, in Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, 2001; and Ausubel, F. et al., (Eds.), Short Protocols in Molecular Biology, Wiley, 2002.
  • An example of high stringency hybridization conditions is hybridization of nucleic acids over about 100 nucleotides in length in a solution containing 6 ⁇ SSC, 5 ⁇ Denhardt's solution, 30% formamide, and 100 micrograms/ml denatured salmon sperm at 37° C. overnight followed by washing in a solution of 0.1 ⁇ SSC and 0.1% SDS at 60° C. for 15 minutes.
  • SSC is 0.15M NaCl/0.015M Na citrate.
  • Denhardt's solution is 0.02% bovine serum albumin/0.02% FICOLL/0.02% polyvinylpyrrolidone.
  • SEQ ID NO:1 and SEQ ID NO:3 will hybridize to the complement of substantially identical targets and not to unrelated sequences.
  • Methods of treating a neurological condition in a subject in need thereof are provided according to aspects of the present invention which include delivering a therapeutically effective amount of NeuroD1 to glial cells of the central nervous system or peripheral nervous system of the subject, the therapeutically effective amount of NeuroD1 in the glial cells results in a greater number of neurons in the subject compared to an untreated subject having the same neurological condition, whereby the neurological condition is treated.
  • the conversion of reactive glial cells into neurons also reduces neuroinflammation and neuroinhibitory factors associated with reactive glial cells, thereby making the glial scar tissue more permissive to neuronal growth so that neurological condition is alleviated.
  • neurological condition refers to any condition of the central and/or peripheral nervous system of a subject which is alleviated, ameliorated or prevented by additional neurons. Injuries or diseases which result in loss or inhibition of neurons and/or loss or inhibition of neuronal function are neurological conditions for treatment by methods according to aspects of the present invention.
  • Injuries or diseases which result in loss or inhibition of glutamatergic neurons and/or loss or inhibition of glutaminergic neuronal functions are neurological conditions for treatment by methods according to aspects of the present invention. Loss or inhibition of other types of neurons, such as GABAergic, cholinergic, dopaminergic, norepinephrinergic, or serotonergic neurons can be treated with the similar method.
  • injuries or diseases which result in loss or inhibition of neurons and/or loss or inhibition of neuronal functions including, but not limited to, Alzheimer' s disease, Parkinson disease, Amyotrophic lateral sclerosis (ALS), stroke, epilepsy, physical injury such as brain or spinal cord injury, and tumor, are neurological conditions for treatment by methods according to aspects of the present invention.
  • Alzheimer' s disease Parkinson disease
  • Amyotrophic lateral sclerosis ALS
  • stroke epilepsy
  • physical injury such as brain or spinal cord injury
  • tumor are neurological conditions for treatment by methods according to aspects of the present invention.
  • a therapeutically effective amount is intended to mean an amount of an inventive composition which is effective to alleviate, ameliorate or prevent a symptom or sign of a neurological condition to be treated.
  • a therapeutically effective amount is an amount which has a beneficial effect in a subject having signs and/or symptoms of a neurological condition.
  • treat refers to alleviating, inhibiting or ameliorating a neurological condition, symptoms or signs of a neurological condition, and preventing symptoms or signs of a neurological condition, and include, but are not limited to therapeutic and/or prophylactic treatments.
  • Combinations of therapies for a neurological condition of a subject are administered according to aspects of the present invention.
  • an additional pharmaceutical agent or therapeutic treatment administered to a subject to treats the effects of disruption of normal blood flow in the CNS in an individual subject in need thereof include treatments such as, but not limited to, removing a blood clot, promoting blood flow, administration of one or more anti-inflammation agents, administration of one or more anti-oxidant agents, and administration of one or more agents effective to reduce excitotoxicity
  • subject refers to humans and also to non-human mammals such as, but not limited to, non-human primates, cats, dogs, sheep, goats, horses, cows, pigs and rodents, such as but not limited to, mice and rats; as well as to non-mammalian animals such as, but not limited to, birds, poultry, reptiles, amphibians.
  • non-human mammals such as, but not limited to, non-human primates, cats, dogs, sheep, goats, horses, cows, pigs and rodents, such as but not limited to, mice and rats; as well as to non-mammalian animals such as, but not limited to, birds, poultry, reptiles, amphibians.
  • inventive compositions and methods are illustrated in the following examples. These examples are provided for illustrative purposes and are not considered limitations on the scope of inventive compositions and methods.
  • Wild type (WT) FVB/NJ and GFAP-GFP transgenic mice were employed for the majority of the experiments described in this study.
  • GFAP-GFP transgenic mice were purchased from the Jackson Laboratory [FVB/N-Tg (GFAPGFP)14Mes/J], described in Zhuo, L. et al., 1997, Dev Biol 187, 36-42, and mated with FVB/NJ mice (Jackson Laboratory).
  • Endothelin-1 (1-31) was injected into the motor cortex of the adult WT FVB/NJ or GFAPA-GFP transgenic mice (28-40 g, 5-10 months old) to produce focal ischemic injury as described in Horie, N. et al., 2008, J. Neurosci. Methods 173, 286-290; and Roome, R. B. et al., 2014, J. Neurosci. Methods, 233, 34-44.
  • mice Under anesthesia by intra-peritoneal injection of ketamine/xylazine (100 mg/kg ketamine; 12 mg/kg xylazine), mice were placed in a stereotaxic apparatus with the skull and bregma exposed by a midline incision. A small hole of ⁇ 1 mm was drilled in the skull at the coordinates of the forelimb motor cortex (relative to the bregma): +0.2 mm anterior-posterior (AP), ⁇ 1.5 mm medial-lateral (ML). ET-1 (1-31) (Peptide international, Inc., PED-4360-s) was dissolved at 2 ⁇ g/ ⁇ l in phosphate-buffered saline (PBS; OSM ⁇ 320, pH ⁇ 7.3).
  • PBS phosphate-buffered saline
  • a total volume of 0.5 ⁇ l (1 ⁇ g) was injected into each site starting from ⁇ -1.6 mm dorsal-ventral (DV).
  • the injection was performed by infusion pump throughout 10 min and the injection needle was withdrawn slowly at 0.1 mm/min. After injection, the needle was kept at 1.1 mm DV for additional 3 min before fully withdrawn.
  • Viral injection followed a similar procedure, with the exception being that viral injection was typically performed around 10 days after ET-1 injection through the same hole drilled for ET-1 injection.
  • mice 5-10 months old mice were used. Mice were housed in a 12 hr light/dark cycle and supplied with sufficient food and water. Both male and female mice were used, except only male mice were used in the behavioral experiments.
  • the hGFAP promoter was obtained from pDRIVE-hGFAP plasmid (InvivoGen, Inc.) and inserted into pAAV-MCS (Cell Biolab) between MluI and SacII to replace the CMV promoter.
  • the Cre gene was obtained by PCR from hGFAP-Cre (Addgene plasmid #40591) and inserted into pAAV MCS between EcoRI and Sall sites to generate pAAV-hGFAP::Cre vector.
  • cDNAs coding NeuroD1, mCherry or GFP were obtained by PCR using the retroviral constructs described in Guo, Z. et al., 2014, Cell Stem Cell 14, 188-202.
  • the NeuroD1 gene were fused with P2A-mCherry or P2A-GFP and subcloned into the pAAV-FLEX-GFP vector (Addgene plasmid # 28304) between Kpn1 and Xho 1 sites. Plasmid constructs were sequenced for verification.
  • Recombinant AAV9 was produced in 293AAV cells (Cell Biolabs). Briefly, polyethylenimine (PEI, linear, MW 25,000) was used for transfection of triple plasmids: the pAAV expression vector; pAAV9-RC (Cell Biolab); and pHelper (Cell Biolab).
  • PEI polyethylenimine
  • Virus titers were 1.2 ⁇ 10 12 GC/ml for hGFAP::Cre, hGFAP::NeuroD1-GFP and hGFAP::GFP, 1.4 ⁇ 10 12 GC/ml for CAG::FLEX-NeuroD1-P2A-GFP and CAG::FLEX-NeuroD1-P2A-mCherry, and 1.6 ⁇ 10 12 GC/ml for CAG::FLEX-mCherry-P2A-mCherry and CAG::FLEX-GFP-P2A-GFP, determined by QuickTiterTM AAV Quantitation Kit (Cell Biolabs).
  • the pCAG-NeuroD1-IRES-GFP and pCAG-GFP were described in Guo, Z. et al., 2014, Cell Stem Cell 14, 188-202.
  • gpg helper-free human embryonic kidney (HEK) cells were transfected with the target plasmid together with vesicular stomatitis virus glycoprotein (VSV-G) vector to produce the retroviruses expressing NeuroD1 or GFP.
  • VSV-G vesicular stomatitis virus glycoprotein
  • the titer of retroviral particles was about 10 7 particles/ml, determined after transduction of HEK cells.
  • Brain sections were permeabilized in 2% Triton X-100 in PBS for 1 hr, followed by incubation in blocking buffer (2.5% normal goat serum, 2.5% normal donkey serum, and 0.1% Triton X-100 in PBS) for 1 hr.
  • the primary antibodies were added into the blocking buffer with brain sections and incubated overnight at 4° C.
  • brain sections were incubated with secondary antibodies conjugated with different fluorophores (1:800, Jackson ImmunoResearch) for 1 hr at room temperature, washed in Triton-PBS, and then mounted onto a glass slide with an anti-fading mounting solution containing DAPI (Invitrogen). Images were acquired with confocal microscopes (Olympus FV1000 or Zeiss LSM800) and a Keyence microscope.
  • the primary antibody was withdrawn and only a secondary antibody was used for immunostaining as a side-by-side control for all the primary antibodies. No specific signal was detected in these controls.
  • mice Deeply anesthetized mice were perfused with artificial cerebral spinal fluid as described above, followed by 15 ml 0.5 mg/ml Sulfo-NHS-LC-Biotin in PBS.
  • brain sections were incubated with Texas Red Streptavidin (Vector SA-5006), 1:800 diluted in PBS+0.3% triton+2.5% normal goat or donkey serum at room temperature for 1 hr, followed by normal mounting procedures.
  • mice were anaesthetized with 2.5% avertin, and then perfused with NMDG-based cutting solution (in mM): 93 NMDG, 93 HCl, 2.5 KCl, 1.25 NaH 2 PO 4 , 30 NaHCO 3 , 20 HEPES, 15 glucose, 12 N-Acetyl-L-cysteine, 5 sodium ascorbate, 2 thiourea, 3 sodium pyruvate, 7 MgSO 4 , 0.5 CaCl 2 , pH 7.3-7.4, 300 mOsmo, bubbled with 95% O 2 /5% CO 2 .
  • NMDG-based cutting solution in mM
  • Coronal sections of 300 ⁇ m thickness were cut around AAV-injected cortical areas with a vibratome (VT1200S, Leica, Germany) at room temperature. Slices were collected and incubated at 33.0 ⁇ 1.0° C. in oxygenated NMDG cutting solution for 10-15 minutes. Slices were then transferred to holding solutions with continuous 95% O 2 /5% CO 2 bubbling (in mM): 92 NaCl, 2.5 KCl, 1.25 NaH 2 PO 4 , 30 NaHCO 3 , 20 HEPES, 15 glucose, 12 N-Acetyl-L-cysteine, 5 sodium ascorbate, 2 Thiourea, 3 sodium pyruvate, 2 MgSO 4 , 2 CaCl 2 .
  • a single slice was transferred to the recording chamber continuously perfused with standard aCSF (artificial cerebral spinal fluid) saturated by 95% O 2 /5% CO 2 at 33.0 ⁇ 1.0° C.
  • the standard aCSF contained (in mM): 124 NaCl, 2.5 KCl, 1.25 NaH 2 PO 4 , 26 NaHCO 3 , 10 glucose, 1.3 MgSO 4 , 2.5 CaCl 2 .
  • pipette solution contained (in mM): 120 Cs-Methanesulfonate, 10 KCl, 10 Na-phosphocreatine, 10 HEPES, 5 QX-314, 1 EGTA, 4 MgATP and 0.3 Na 2 GTP, pH 7.3 adjusted with KOH, 280-290 mOsm.
  • sEPSCs spontaneous excitatory postsynaptic currents
  • sIPSCs spontaneous inhibitory postsynaptic currents
  • pipette solution contained (in mM): 120 Cs-Methanesulfonate, 10 KCl, 10 Na-phosphocreatine, 10 HEPES, 5 QX-314, 1 EGTA, 4 MgATP and 0.3 Na 2 GTP, pH 7.3 adjusted with KOH, 280-290 mOsm.
  • biocytin was added to the pipette solution.
  • the cell membrane potentials were held at ⁇ 70 mV (the reversal potential of GABAA receptors) for sEPSC recording, and 0 mV (the reversal potential of ionotropic glutamate receptors) for sIPSC recording, respectively.
  • Data were collected with a MultiClamp 700A amplifier and analyzed with pCLAMP10 software (Molecular Devices).
  • ET-1 (1-31) was injected into two points of the forelimb motor cortex in order to induce severe motor deficits.
  • the coordinates of the two points are: (i) +0.2 mm AP, +/ ⁇ 1.35 mm ML; (ii) +0.38 mm AP, +/ ⁇ 2.45 mm ML (+or ⁇ ML was based on the opposite side of the dominant forelimb in pre-stroke pellet retrieval training).
  • the food pellet retrieval chamber was modified from the staircase equipment for mice described in Baird, A. L. et al., 2001, Brain Res. Bull. 54, 243-250; and Roome, R. B. et al., 2014, J. Neurosci. Methods, 233, 34-44.
  • the staircase was changed to an arm with a single V-shape well and a small baffle at the end to prevent food pellet dropped outside the well during the test.
  • the well is 33 mm long and the lowest part of the well is 16 mm from the bottom of top board, as illustrated in the FIG. 14A .
  • mice were food-deprived for 18 hrs to increase their motivation.
  • mice were given 30 sucrose pellets (14 mg, TestDiet, Inc) in home cages for familiarization.
  • sucrose pellets 14 mg, TestDiet, Inc
  • the top board of the center plinth was removed to reduce difficulty and encourage animals to try reaching the food.
  • Five pellets were placed in the well either on the right side or left side to determine the dominant forelimb. Mice were given 5 min and tested twice on each side. The dominant side was determined at this stage by counting how many pellets were retrieved. Most animals could get -5 pellets on one side and sometimes both sides. If the animals failed to get any pellet, they were excluded from further tests.
  • the top board was placed back onto the center plinth and 8 pellets were placed in the well at the dominant side. Animals were tested 3 consecutive times every day, each time being given 5 min for pellet retrieval. The animals would be used for further surgery and testing if the average retrieval was more than 4 pellets on three consecutive training days.
  • ischemic stroke was induced in the contralateral motor cortex controlling the dominant forelimb.
  • a pellet retrieval test was performed to determine their functional levels. If the number of pellets retrieved were less than half of their pre-stroke level, these animals would be used for viral injection the next day (10 days post stroke).
  • pellet retrieval tests were performed at 20, 30, 50, and 70 dps to assess their functional recovery. For each time point, pellet retrieval tests were repeated on two consecutive days, with day 1 as training and day 2 as the actual test. Animals were coded and a second experimenter who was blind to the identity of the animals performed the test and pellet counting.
  • a 1 cm 2 grid wire mesh of 24 cm long and 20 cm wide, elevated to 24 cm high was used.
  • a video camera was placed below the grid, facing upwards with a 45° angle. Slow motion video footages were recorded to assess the animals' foot faults.
  • An individual mouse was placed on top of the grid floor and allowed to freely walk for 5 min. The animals were coded and video footage was analyzed offline by a second researcher who was blind to the animal identity.
  • Foot fault percentage was calculated as: the number of foot faults/total number of steps ⁇ 100.
  • the cylinder test was based on methods described in Roome, R. B. et al., 2014, J. Neurosci. Methods, 233, 34-44. This test involved video recording of mice rising up and touching the sidewall of a transparent cylinder with forelimbs (10 cm diameter, 15 cm tall). Each animal was placed into the cylinder on a transparent board and video-recorded with a camera from below. The experiment was stopped after the mouse attempted to rise and touch the sidewall for >30 times, which normally lasted -3-4 minutes.
  • a normal paw touch is defined as the animal placing both paws on the wall while rising, and using both paws to push against the wall while descending.
  • An abnormal touch involves only using one of the paws to touch the wall, or one paw dragging along the wall after touching.
  • Dragging behavior is defined as one paw, typically the injured one, sliding along the sidewall without holding steady against the wall.
  • a non-touching behavior is defined as the mouse not using its injured paw but only using the non-injured paw while rising and descending. Non-touching is considered a more severe injury phenotype since the mouse totally abandons the injured paw.
  • the normal rising and touching behavior was quantified as: normal paw touches/total attempts ⁇ 100.
  • the cortical tissues around the injury core (approximately 2 mm ⁇ 2 mm square) were taken after perfusion and flash-frozen in liquid nitrogen.
  • the RNA extraction was conducted using Macherey-Nagel NucleoSpin RNA kit and the RNA concentration was measured by NanoDrop.
  • the cDNAs were synthesized using Quanta Biosciences qScript cDNA supermix.
  • the reaction mixture was incubated at 25° C. for 5 min, 42° C. for 30 min, 85° C. for 5 min and held at 4° C.
  • the mixture was then diluted 5-fold with RNase/DNase-free water and 2.5 ⁇ l was taken for qRT-PCR.
  • the primers for qRT-PCR were designed using Applied Biosystems Primer Express software.
  • the qRT-PCR was conducted using Quanta Biosciences PerfeCTa SYBR Green Supermix, ROX. GAPDH was used as an internal control and non-stroke cortical tissue from healthy mice was used as control. Comparative Ct method was used for calculation of fold change and Prism 6 was used for statistical analysis and bar graphs.
  • the slice Golgi kit (Bieoenno Tech, LLC) was used for Golgi staining.
  • mice were perfused with aCSF, followed by fixative from the kit.
  • the brains were removed and impregnated for five days, sectioned by vibratome at 200 ⁇ m, stained and mounted based on the kit protocol, coverslipped with DPX mountant.
  • Brightfield images at 4 ⁇ and 100 ⁇ were taken using a Keyence BZ-9000 Fluorescent microscope.
  • Cortical areas were quantified using the images taken with a 4 ⁇ lens in a Keyence BZ-9000 Fluorescent microscope. Three slices around the injection point (+0.2 AP) with most obvious stroke injury were chosen for image-taking. DAPI and NeuN signals were used to identify the cortical upper boundary and the lower interface with cingulate cortex. Cortical areas from the midline to 3 mm lateral were measured by ImageJ.
  • GFP NeuroD1-GFP
  • cortical neuron marker Tbr1, Emx 1, Satb2, PV, GABA
  • NeuN and PV cell number was counted on the images of NeuN or PV immunostaining taken by the tile function of Zeiss LSM800 confocal microscope within the cortical areas from 500 ⁇ m to 2500 ⁇ m lateral of the midline.
  • the confocal software Zen was used for cell counting. Slices around the injection point (+0.2 AP) with most obvious stroke injury were used for image taken.
  • confocal images of NeuN immunostaining were used for quantification at 17 dpi. Three images were taken at virus-infected areas close to the stroke infarct (within 500 ⁇ m from stroke core boarder), as shown in FIG. 4A . Z stack function was used with 10 sections at 1 ⁇ m interval. The confocal software Zen was used for cell counting. For NeuroD1 group, cells positive for both GFP (NeuroD1-GFP) and NeuN (converted neurons) or NeuN positive GFP negative (pre-existing neurons) were counted. For GFP control group, total number of NeuN positive cells were counted.
  • the sEPSCs and sIPSCs were analyzed using Mini Analysis Program (Synaptosoft, New Jersey, USA). To avoid multiple detections of large events, the analysis results were further checked visually after auto-detection. To compare sEPSCs and sIPSCs frequency between different groups, over 200 events or at least 3-min recording periods were sampled for each cell before obtaining the average value.
  • Ly6C number quantification single-layer 40 ⁇ images were taken by Zeiss confocal.
  • the Ly6C intensity was measured by ImageJ and blood vessels having intensities three times stronger than background were identified as positive signals.
  • the number of Ly6C+blood vessels in three images in peri-infarct areas was quantified and averaged for each mouse.
  • ET-1 (1-31) produced more severe and prolonged brain damage than ET-1 (1-21) (see, FIG. 2A ).
  • a genetic strain of mice with FVB background gave more severe stroke damage than the commonly used B6/C57 mice (see, FIG. 2A ).
  • significant cortical tissue loss was observed with injection of ET-1 (1-31) into the motor cortex of FVB mice (see, FIG. 1A and FIG. 1B )—establishing a severe focal stroke model with consistent tissue damage over a time course of 2 months.
  • a suitable time window for NeuroD1 viral injection after stroke was determined. Because NeuroD1 can convert reactive astrocytes into neurons, the stroke areas were surveyed at different time points to examine when astrocytes became reactive.
  • FIG. 1C Five days post stroke (dps), there was a significant loss of neuronal signal NeuN, as expected (see, FIG. 1C ). Interestingly, the GFAP signal, a commonly used reactive astrocyte marker, was also very low, suggesting that the astrocytes had not been activated at this early stage. At 10 dps, however, the GFAP signal was significantly upregulated (see, FIG. 1D ), indicating that the astrocytes became reactive around this time.
  • retroviruses While retroviruses have the advantage of mainly targeting the dividing reactive glial cells after injury, they cannot generate a large number of neurons for neural repair because the number of dividing glial cells is rather limited. In order to generate a sufficient number of neurons for functional repair after stroke, adeno-associated virus (AAV) was used to infect both dividing and non-dividing glial cells. AAV has the advantage of high infection rate and low pathogenicity in humans, and has been approved by FDA for clinical trials in the treatment of CNS disorders.
  • AAV adeno-associated virus
  • an AAV vector (recombinant serotype AAV9) expressing NeuroD1 under the control of a human GFAP promoter (hGFAP::NeuroD 1-P2A-GFP) was constructed. After two weeks of infection, many NeuroD1-infected cells were also converted into NeuN-positive neurons (see, FIG. 1F ). However, one caveat with the GFAP promoter-driven expression is that the GFAP promoter will be silenced after astrocyte-to-neuron conversion, making it difficult to distinguish converted from non-converted neurons.
  • the GFAP promoter and NeuroD1 were separated into two different AAV9 vectors by using the Cre-FLEX (flip-excision) homologous recombination system (see, Atasoy et al., 2008).
  • a human GFAP promoter was then used to drive the expression of Cre recombinase (hGFAP::Cre), which could then act on two pairs of heterotypic, antiparallel 1oxP-type recombination sites flanking an inverted sequence of NeuroD1-P2A-GFP (or NeuroD1-P2A-mCherry) under the control of a CAG promoter in a separate vector (CAG::FLEX-NeuroD1-P2A-GFP/mCherry) (see, FIG. 2B ).
  • CAG::FLEX-NeuroD1-P2A-GFP/mCherry see, FIG. 2B .
  • the GFAP signal showed a dramatic reduction from 4 to 17 dpi (see, FIG. 3A , bottom row), suggesting that many AAV-NeuroD1-infected cells lost astrocytic identity.
  • NeuN staining revealed very few neurons among the mCherry-infected control group, but the NeuroD 1-mCherry expressing cells became increasingly colocalized with NeuN (see, FIG. 3B ).
  • an Tri-AAV tracing system was developed to track astrocyte-converted neurons, which included a virus that expressed GFP in the astrocytes (AAV9 hGFAP::GFP) together with a Cre-FLEX system of the present invention (see, FIGS. 4A-4C ).
  • AAV9 hGFAP::GFP virus that expressed GFP in the astrocytes
  • Cre-FLEX system of the present invention see, FIGS. 4A-4C .
  • mCherry control group only 10% of GFP+ cells showed NeuN+ signal by 60 dpi; but in NeuroD1-mCherry group, 95% of GFP+ cells were also NeuN+ at 60 dpi (quantified in FIG. 4B ), further confirming the high conversion efficiency of NeuroD1 when expressed in astrocytes.
  • astrocyte-converted neurons were immunopositive for cortical pyramidal neuron markers including Emx1, Tbr1, and Satb2 (see, FIG. 3E and FIG. 3F ). Only about 10% were immunopositive for GABAergic neuron markers including parvalbumin and GABA (see, FIG. 3F ).
  • astrocyte-converted neurons showed similar neuronal identity to their neighboring cortical neurons.
  • astrocytes did show a reduction in the NeuroD1-infected areas, there were still a significant number of astrocytes remaining (see, FIG. 5B ). Surprisingly, not only was the astrocyte number reduced, but astrocyte morphology was significantly changed in the NeuroD1-converted areas. In the control group, astrocytes were hypertrophic and densely intermingled together in the areas surrounding the injury core (see, FIG. 5B , top row); whereas in the NeuroD1 group, most of the astrocytes had elongated processes with only a few showing hypertrophic morphology (see, FIG. 5B , bottom row; and FIG. 6A-C ). suggests that the remaining astrocytes became less reactive in the NeuroD1-converted areas.
  • GFAP-GFP transgenic mice were used to illuminate the entire astrocyte morphology. Similar to GFAP immunostaining, the use of GFAP-GFP mice also revealed fewer hypertrophic astrocytes in the NeuroD1 group (see, FIG. 5C ). Notably, an abundant amount of GFP-labeled astrocytes in the NeuroD1-mCherry expressing areas was observed (see, FIG. 5C , bottom row), suggesting that astrocytes are not depleted after conversion. This differs markedly from the astrocyte-killing approach. The persistence of astrocytes with in vivo cell conversion is likely due to the fact that astrocytes have an intrinsic capacity to divide and regenerate themselves.
  • microglia in the control group were mostly amoeboid type (see, 5 E, top); but in the NeuroD1 group, microglia mainly showed ramified shapes with clear processes (see, FIG. 5E , bottom; quantified in FIG. 6D ; see also FIG. 6A-B for 7 dpi and 40 dpi signal, respectively).
  • CSPGs chondroitin sulfate proteoglycans
  • LCN2 lipocalin-2
  • RT-PCR experiments at 17 dpi were run to quantitatively analyze the glial markers and cytokines in the control or NeuroD1-infected cortical tissues. It was first confirmed that NeuroD1 was indeed highly expressed in the NeuroD1-injected samples (see, FIG. 5H ).
  • the GFAP transcriptional level increased by ⁇ 50-fold after stroke but significantly decreased after NeuroD1 treatment (see, FIG. 5H ).
  • the neuroinflammation related factor LCN2 was drastically increased by ⁇ 1600-fold after stroke but decreased to only 15-fold in NeuroD1-treatment group (see, FIG. 5G ).
  • the pro-inflammatory factors including interferon ⁇ , TNF ⁇ , interleukin 1 ⁇ (IL-1-1 ⁇ ), and interleukin 6 (IL-6) all showed an increase after stroke, but were downregulated after NeuroD1-treatment (see, FIG. 5I ).
  • the unexpected reduction of neuroinflammation after NeuroD1-treatment suggests that converting reactive astrocytes into neurons has a much broader impact than simply generating new neurons.
  • the NeuN+ neurons in the NeuroD1-infected areas more than tripled the ones in the control group.
  • immunostaining of neuronal dendritic markers including microtubule-associated protein 2 (Map2) and SMI32 showed clear dendritic morphology in the NeuroD1 group, but not in the GFP control group (see, FIG. 7D and FIG. 7E ; Map2 quantified in FIG. 8C ; see also FIG. 8B for Map2 staining at 40 dpi).
  • the total number of NeuN-positive neurons within the stroke injured cortical areas 500 ⁇ m to 2500 ⁇ m from midline, injection site at 1500 ⁇ m from midline) from two weeks to two months after viral infection were quantified.
  • the number of neurons in the NeuroD1 group significantly increased over two month period, rescuing ⁇ 70% of the total neurons in the stroke areas (see, FIG. 71 ).
  • FIG. 9A The analysis of the motor cortical tissue revealed that the control group lost 70% of the injured tissue over a 2-month period, whereas NeuroD1-treated group preserved the majority of the injured tissue with only 20% tissue loss (see, FIG. 9B ; see also FIG. 10A for serial sections).
  • H&E hematoxylin and eosin staining at 60 dpi (see, FIG. 10B ) was also performed.
  • the control group besides tissue damage, many neurons appeared shrunken with eosinophilic cytoplasm and pyknotic nuclei; while in the NeuroD1 group, neurons showed much healthier morphology.
  • axon bundles in the striatum originating from the NeuroD 1-converted neurons in the stroke areas were detected.
  • NeuroD1-infected neurons in the stroke areas projected to after in vivo cell conversion, through sagittal section analysis it was determined that the cortical neurons converted by NeuroD1 could send axons to striatum, thalamus, and hypothalamus ipsilaterally (see, FIG. 9C ). Contralaterally, NeuroD1-converted neurons projected their axons across the midline through corpus callosum to the contralateral cortical regions (see, FIG. 10C ). Therefore, the NeuroD1-converted neurons can be integrated into global brain circuits.
  • Neuronal functions in the stroke areas after NeuroD 1-mediated cell conversion were also investigated.
  • Whole-cell patch-clamp recordings were performed on AAV-NeuroD1-GFP-infected neurons in the cortical slices at 2 months after viral injection (see, 9 D). Injecting depolarizing currents into the neurons triggered repetitive action potentials (see, FIG. 9D ).
  • FIG. 9E demonstrates that the neuron recorded (biocytin positive) was indeed NeuroD 1-positive.
  • the biocytin immunostaining also illuminated the complex dendritic structures of the NeuroD1-converted neurons in the motor cortex (see, FIG. 9E ).
  • NeuroD1-treatment can rescue brain tissue loss and form neural circuits in the stroke areas, can blood vessels and blood-brain-barrier be restored after cell conversion?
  • FIGS. 11A-11H blood vessels and astrocytic endfeet that contact blood vessels to form blood-brain-barrier in the stroke areas were examined (see, FIGS. 11A-11H ).
  • ET-1 1-31)-induced focal stroke, significant disruption of both blood vessels and astrocytic endfeet were observed (see, FIG. 11A , bottom row).
  • the AQP4-labeled astrocytic endfeet were detached from blood vessels and became co-localized with GFAP (see, FIG. 11A , bottom row)—suggesting a mislocalization of AQP4 from the endfeet to other areas of astrocytes.
  • FIG. 11B-C After NeuroD1-treatment, a significant restoration of blood vessels together with re-association of AQP4-labeled astrocytic endfeet in the stroke areas was observed (see, FIG. 11B-C ; 17 dpi, 27 dps).
  • Quantitative analysis showed a significant decrease of both AQP4 intensity (see, FIG. 11F ) and AQP4-covered areas (see, FIG. 11G ) in NeuroD1-treated group compared to the GFP control group. Even in NeuroD1 group, the AQP4 signal appeared more associated with blood vessels in the regions where NeuroD1 was highly expressed than the areas with less NeuroD1 signal (see, FIG. 12G ).
  • the blood vessels showed a significant increase in the NeuroD1 group compared to the control group (see, FIG. 11H ).
  • BBB blood brain barrier
  • a biotin perfusion test was used to detect BBB leakage.
  • biotin showed strong signal outside the blood vessel (labeled by Ly6C) in control group, indicating BBB leakage (see, FIG. 12H ).
  • ET-1 (1-31) was injected at two points on one cortical side (one at forelimb motor cortex and one at forelimb sensory cortex) to produce a severe unilateral stroke on the dominant side according to the food pellet retrieval test.
  • mice were food-deprived before the test to increase their motivation for food.
  • normal animals were trained to retrieve on average 5-6 pellets out of 8 total in 5 minutes (see, FIG. 14A for the pellet retrieval device).
  • a cylinder test that assesses the forelimb function when animals rise and touch the sidewall of a cylinder was also performed. Normal animals typically use both forelimbs to touch and push steadily against the sidewall and normal touching rate is ⁇ 85%. After a unilateral stroke in the forelimb motor cortex, the function of the contralateral forelimb was significantly weakened and the impaired limb often failed to touch the sidewall or dragged along the wall after briefly touching (see, FIG. 13D , normal touching rate dropped to ⁇ 35%).
  • the NeuroD1 group showed a significant recovery in touching the sidewall with both forelimbs ( ⁇ 65%), whereas the majority of the GFP control mice remained weak and unable to use the injured forelimb (see, FIG. 13D ).
  • injection of PBS as a sham control for ET-1 (1-31) did not result in any behavioral deficits, and injection of ET-1 (1-31) without viral injection produced similar deficits as the GFP control viral injection.
  • the non-stroke side forelimb did not show significant deficits compared to the control animals without stroke.
  • the forelimb preferentially used to take the food pellets during the training sessions was stroke injured and tested.
  • the mice were sacrificed and anatomical and immunocytochemical analysis were performed. Consistent with the behavioral deficits, the brains of the GFP control group displayed a remarkable cavity in the stroke areas, whereas the NeuroD1 group showed much smaller injury at the stroke site (see, FIG. 14B ).
  • both glial morphology and function were significantly improved in NeuroD1-infected regions.
  • the significant number of astrocytes after neural conversion is consistent with the proliferation capability of reactive astrocytes.
  • NeuroD1-treatment results in the repair of blood vessels and restoration of BBB in the stroke areas.
  • This type of repair after neuroregeneration is consistent with neurovascular interaction during brain development and neural repair.
  • NeuroD 1-mediated in vivo cell conversion has a broad impact on local neuron-glia circuitry, including the generation of a large number of new neurons, decreasing the same number of reactive astrocytes, reducing neuroinflammation, repairing blood vessels, and restoring the BBB.
  • Such landscape change of the entire neuron-glia circuitry is unprecedented.
  • In vivo glia-to-neuron conversion can also functionally rescue the motor deficits caused by ischemic stroke in rodent models. This functional rescue is based on structural changes including both cellular conversion and circuit rebuilding after NeuroD1-treatment. A mixture of NeuroD1-GFP-labeled neurons together with non-labeled neurons in the stroke areas after NeuroD1-treatment was observed. This suggests that the repaired brain circuitry is an integration of both newly generated neurons and old mature neurons.
  • NeuroD1-converted neurons are mainly glutamatergic
  • a significant number of GABAergic neurons that are not labeled by NeuroD1-GFP were observed—suggesting that GABAergic neurons are among the neurons rescued by NeuroD1-treatment.
  • the preservation of GABAergic neurons suggests that NeuroD1-treatment may keep an excitation-inhibition balance after in vivo cell conversion, which is important to achieve normal brain function. Functional recovery is also achieved since NeuroD1-treatment can rescue over 70% of neurons in stroke injured motor cortex.
  • Item 1 A method of treating the effects of disruption of normal blood flow in the CNS in an individual subject in need thereof, comprising:
  • Item 2 The method of item 1, wherein administering exogenous NeuroD1 comprises delivering an expression vector comprising a nucleic acid encoding NeuroD1 to the area.
  • Item 3 The method of item 1 or 2, wherein administering exogenous NeuroD1 comprises delivering a recombinant viral expression vector comprising a nucleic acid encoding NeuroD1 to the area.
  • Item 4 The method of any of items 1-3, wherein expressing exogenous NeuroD1 comprises delivering a recombinant adeno-associated virus expression vector comprising a nucleic acid encoding NeuroD1 to the area.
  • Item 5 The method of any of items 1-4, wherein expressing exogenous NeuroD1 comprises delivering an effective “flip-excision” recombinant expression vector combination of 1) a recombinant adeno-associated virus expression vector comprising a nucleic acid encoding NeuroD1 and 2) a recombinant adeno-associated virus expression vector comprising a nucleic acid encoding a site-specific recombinase to the area.
  • Item 6 The method of any of items 1-5, wherein NeuroD1 is the only exogenously expressed transcription factor delivered to the area.
  • Item 7 The method of any of items 1-6, wherein exogenous NeuroD1 is administered at a time following disruption of normal blood flow in the CNS when reactive astrocytes are present.
  • Item 8 The method of any of items 1-7, wherein exogenous NeuroD1 is administered at a time following disruption of normal blood flow in the CNS when a glial scar is present.
  • Item 9 The method of any of items 1-8, wherein the disruption of normal blood flow in the CNS is due to a disorder selected from the group consisting of: ischemia, thrombosis, embolism, hemorrhage, chronic disease mediated restriction of blood vessels and a combination of any two or more thereof.
  • Item 10 The method of any of items 1-9, wherein the disruption of normal blood flow in the CNS is due to a disorder selected from the group consisting of: ischemic stroke; hemorrhagic stroke; brain aneurysm; concussion, tumor, infection, inflammation, traumatic brain injury, traumatic spinal injury; ischemic or hemorrhagic myelopathy (spinal cord infarction); global ischemia as caused by cardiac arrest or severe hypotension (shock); hypoxic-ischemic encephalopathy as caused by hypoxia, hypoglycemia, or anemia; CNS embolism as caused by infective endocarditis or atrial myxoma; fibrocartilaginous embolic myelopathy; CNS thrombosis as caused by pediatric leukemia; cerebral venous sinus thrombosis as caused by nephrotic syndrome (kidney disease), chronic inflammatory disease, pregnancy, use of estrogen-based contraceptives, meningitis, dehydration; or a combination
  • administering the therapeutically effective dose of NeuroD1 comprises administering a recombinant expression vector comprising a nucleic acid sequence encoding NeuroD1 protein, wherein the nucleic acid sequence encoding NeuroD1 protein comprises a nucleic acid sequence selected from the group consisting of: a nucleic acid sequence encoding SEQ ID NO:2 or a functional fragment thereof; a nucleic acid sequence encoding SEQ ID NO:4 or a functional fragment thereof; SEQ ID NO:1 or a functional fragment thereof; SEQ ID NO:3 or a functional fragment thereof; and a nucleic acid sequence encoding a protein which has 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 81%, 82%, 83%, 84%, 85%, 86%,87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%,
  • Item 12 The method of any of items 1-11, wherein administering the therapeutically effective dose of NeuroD1 comprises stereotactic injection in or near an injury site.
  • Item 13 The method of any of items 1-12, wherein the nucleic acid sequence encoding NeuroD1 is operably linked to a ubiquitous promoter.
  • Item 14 The method of any of items 5-12, wherein the nucleic acid sequence encoding a site-specific recombinase is operably linked to a glial-cell specific promoter.
  • Item 15 The method of item 14, wherein the glial-cell specific promoter is a GFAP promoter.
  • Item 16 The method of item 15, wherein the GFAP promoter is a human GFAP promoter.
  • Item 17 The method of any of items 1-16, further comprising assessing the effectiveness of the treatment in the subject.
  • Item 18 The method of item 17, wherein assessing the effectiveness of the treatment in the subject comprises an assay selected from: electrophysiology assay, blood flow assay, tissue structure assay, function assay and a combination of any two or more thereof.
  • Item 19 The method of item 17, wherein assessing the effectiveness of the treatment in the subject comprises an electroencephalogram.
  • Item 20 The method of item 17, wherein assessing the effectiveness of the treatment in the subject comprises an assay of blood flow selected from the group consisting of: Near Infrared Spectroscopy and fMRI.
  • Item 21 The method of item 17, wherein assessing the effectiveness of the treatment in the subject comprises an assay of tissue structure selected from the group consisting of: MRI, PET scan, CAT scan and ultrasound.
  • Item 22 The method of item 17, wherein assessing the effectiveness of the treatment in the subject comprises a behavioral assay.
  • Item 23 The method of item 17, wherein assessing the effectiveness of the treatment in the subject comprises an assay performed prior to administration of the therapeutically effective dose of exogenous NeuroD1.
  • Item 24 The method of any of items 1-23, wherein 1-500 ⁇ l of a pharmaceutically acceptable carrier containing adeno-associated virus particles comprising a nucleic acid encoding NeuroD1 at a concentration of 10 10 -10 14 adeno-associated virus particles/ml carrier is injected into the subject, in the area where normal blood flow has been disrupted, at a controlled flow rate of 0.1-5 ⁇ l/min.
  • Item 25 The method of any of claims 1 - 24 , further comprising a treatment selected from the group consisting of: removing a blood clot, promoting blood flow, administering an anti-inflammatory, administering an anti-oxidant, reducing excitotoxicity, and two or more thereof.
  • a composition comprising 1) a recombinant adeno-associated adenovirus expression vector comprising a glial cell specific promoter operably linked to a nucleic acid encoding a site-specific recombinase and 2) a recombinant adeno-associated adenovirus expression vector comprising a ubiquitous promoter operably linked to a nucleic acid encoding NeuroD1, wherein the nucleic acid encoding NeuroD1 is inverted and flanked by two sets of site-specific recombinase recognition sites such that action of the recombinase irreversibly inverts the nucleic acid encoding NeuroD1 such that NeuroD1 is expressed in a mammalian cell.
  • Item 27 A method of treating the effects of disruption of normal blood flow in the CNS in an individual subject in need thereof substantially as described herein.
  • Item 28 A flip-excision expression vector combination substantially as described herein.
  • SEQ ID NO: 1 Human NeuroD1 nucleic acid sequence encoding human NeuroD1 protein - 1071 nucleotides, including stop codon ATGACCAAATCGTACAGCGAGAGTGGGCTGATGGGCGAGCCTCAGCCCCAAGGTCCTCCAAGCT GGACAGACGAGTGTCTCAGTTCTCAGGACGAGGAGCACGAGGCAGACAAGAAGGAGGACGACCT CGAAGCCATGAACGCAGAGGAGGACTCACTGAGGAACGGGGGAGAGGAGGAGGACGAAGATGAG GACCTGGAAGAGGAGGAAGAAGAGGAAGGAGGATGACGATCAAAAGCCCAAGAGACGCGGCC CCAAAAAGAAGAAGATGACTAAGGCTCGCCTGGAGCGTTTTAAATTGAGACGCATGAAGGCTAA CGCCCGGGAGCGGAACCGCATGCACGGACTGAACGCGGCGCTAGACAACCTGCGCAAGGTGGTG CCTTGCTATTCTAAGACGCAGAAGCTGTCCAAAATCGAGACT
  • compositions and methods described herein are presently representative of preferred embodiments, exemplary, and not intended as limitations on the scope of the invention. Changes therein and other uses will occur to those skilled in the art. Such changes and other uses can be made without departing from the scope of the invention as set forth in the claims.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Zoology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biophysics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Epidemiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Immunology (AREA)
  • Plant Pathology (AREA)
  • Microbiology (AREA)
  • Marine Sciences & Fisheries (AREA)
  • Virology (AREA)
  • Toxicology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Magnetic Resonance Imaging Apparatus (AREA)
US16/487,345 2017-02-28 2018-02-28 Regenerating functional neurons for treatment of neural injury caused by disruption of blood flow Abandoned US20200054711A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/487,345 US20200054711A1 (en) 2017-02-28 2018-02-28 Regenerating functional neurons for treatment of neural injury caused by disruption of blood flow

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201762464469P 2017-02-28 2017-02-28
US201762518914P 2017-06-13 2017-06-13
US16/487,345 US20200054711A1 (en) 2017-02-28 2018-02-28 Regenerating functional neurons for treatment of neural injury caused by disruption of blood flow
PCT/US2018/020251 WO2018160712A1 (en) 2017-02-28 2018-02-28 Regenerating functional neurons for treatment of neural injury caused by disruption of blood flow

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/020251 A-371-Of-International WO2018160712A1 (en) 2017-02-28 2018-02-28 Regenerating functional neurons for treatment of neural injury caused by disruption of blood flow

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US18/448,421 Continuation US20240082352A1 (en) 2017-02-28 2023-08-11 Regenerating functional neurons for treatment of neural injury caused by disruption of blood flow

Publications (1)

Publication Number Publication Date
US20200054711A1 true US20200054711A1 (en) 2020-02-20

Family

ID=63370306

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/487,345 Abandoned US20200054711A1 (en) 2017-02-28 2018-02-28 Regenerating functional neurons for treatment of neural injury caused by disruption of blood flow
US18/448,421 Pending US20240082352A1 (en) 2017-02-28 2023-08-11 Regenerating functional neurons for treatment of neural injury caused by disruption of blood flow

Family Applications After (1)

Application Number Title Priority Date Filing Date
US18/448,421 Pending US20240082352A1 (en) 2017-02-28 2023-08-11 Regenerating functional neurons for treatment of neural injury caused by disruption of blood flow

Country Status (5)

Country Link
US (2) US20200054711A1 (ja)
EP (2) EP3589307B1 (ja)
JP (2) JP7364468B2 (ja)
CN (3) CN118384296A (ja)
WO (1) WO2018160712A1 (ja)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20210162003A1 (en) * 2019-10-17 2021-06-03 The Penn State Research Foundation Regenerating functional neurons for treatment of hemorrhagic stroke
WO2024044380A3 (en) * 2022-08-25 2024-04-04 Massachusetts Institute Of Technology Single cell super-resolution proteomics

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4007609A1 (en) * 2019-08-01 2022-06-08 Cornell University Targeted gene therapy to treat neurological diseases
CA3157523A1 (en) * 2019-10-17 2021-04-22 Gong Chen Regenerating functional neurons for treatment of spinal cord injury and als
AU2020368526A1 (en) * 2019-10-17 2022-05-12 The Penn State Research Foundation Regenerating functional neurons for treatment of neurological disorders
WO2021108609A1 (en) * 2019-11-25 2021-06-03 The Penn State Research Foundation Brain repair after traumatic brain injury through neurod1-mediated astrocyte-to-neuron conversion
EP4090754A4 (en) * 2020-01-18 2024-04-24 Aridis Pharmaceuticals, Inc. TUNEABLE TRANSPOSON SYSTEMS
TW202421646A (zh) * 2022-06-30 2024-06-01 日商安斯泰來製藥股份有限公司 腦梗塞之非人類靈長類動物模式之製作方法及腦梗塞治療用醫藥組成物
WO2024143444A1 (ja) * 2022-12-28 2024-07-04 アステラス製薬株式会社 4-アミノピペリジン化合物、その脂質ナノ粒子、及び、それを含む医薬組成物
WO2024162304A1 (ja) * 2023-01-31 2024-08-08 アステラス製薬株式会社 環状アミンを有するカルバモイル脂質又はウレア脂質、それを含む脂質ナノ粒子、及び医薬組成物

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014015261A1 (en) * 2012-07-19 2014-01-23 The Penn State Research Foundation Regenerating functional neurons for treatment of disease and injury in the nervous system

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5648097A (en) 1995-10-04 1997-07-15 Biotek, Inc. Calcium mineral-based microparticles and method for the production thereof
US6391336B1 (en) 1997-09-22 2002-05-21 Royer Biomedical, Inc. Inorganic-polymer complexes for the controlled release of compounds including medicinals
EP1942192A1 (en) * 2007-01-08 2008-07-09 Heinrich-Pette-Institut für experimentelle Virologie und Immunologie Use of a tailored recombinase for the treatment of retroviral infections
CN102858985A (zh) * 2009-07-24 2013-01-02 西格马-奥尔德里奇有限责任公司 基因组编辑方法
CA3087869A1 (en) * 2018-02-02 2019-08-08 The Penn State Research Foundation Methods and materials for treating brain injuries

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014015261A1 (en) * 2012-07-19 2014-01-23 The Penn State Research Foundation Regenerating functional neurons for treatment of disease and injury in the nervous system
US20150250900A1 (en) * 2012-07-19 2015-09-10 The Penn State Research Foundation Regenerating functional neurons for treatment of disease and injury in the nervous system

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Prakash et al, Blood–brain barrier breakdown and neovascularization processes after stroke and traumatic brain injury, Curr. Opin. Neurol. 28(6): 556-564, 2015 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20210162003A1 (en) * 2019-10-17 2021-06-03 The Penn State Research Foundation Regenerating functional neurons for treatment of hemorrhagic stroke
WO2024044380A3 (en) * 2022-08-25 2024-04-04 Massachusetts Institute Of Technology Single cell super-resolution proteomics

Also Published As

Publication number Publication date
EP3589307A4 (en) 2020-01-22
EP3589307A1 (en) 2020-01-08
WO2018160712A8 (en) 2019-08-22
EP4039812A1 (en) 2022-08-10
JP7364468B2 (ja) 2023-10-18
JP2024001178A (ja) 2024-01-09
WO2018160712A1 (en) 2018-09-07
EP3589307B1 (en) 2022-02-16
CN110366423A (zh) 2019-10-22
JP2020514328A (ja) 2020-05-21
US20240082352A1 (en) 2024-03-14
CN118384296A (zh) 2024-07-26
CN118384297A (zh) 2024-07-26

Similar Documents

Publication Publication Date Title
US20240082352A1 (en) Regenerating functional neurons for treatment of neural injury caused by disruption of blood flow
US11167044B2 (en) Regenerating functional neurons for treatment of disease in the nervous system
US20210032300A1 (en) Methods and materials for treating brain injuries
AU2020391479A1 (en) Brain repair after traumatic brain injury through NeuroDl-mediated astrocyte-to-neuron conversion
US20220160825A1 (en) Brain repair after traumatic brain injury through neurod1-mediated astrocyte-to-neuron conversion
US20210162002A1 (en) Regenerating functional neurons for treatment of spinal cord injury and als
US20210162003A1 (en) Regenerating functional neurons for treatment of hemorrhagic stroke
US20210155664A1 (en) Regenerating functional neurons for treatment of neurological disorders

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: THE PENN STATE RESEARCH FOUNDATION, PENNSYLVANIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CHEN, GONG;REEL/FRAME:052153/0916

Effective date: 20191127

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION